Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
ACS Nano ; 18(3): 2485-2499, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38197613

RESUMO

Addressing the critical requirement for real-time monitoring of tumor progression in cancer care, this study introduces an innovative wearable platform. This platform employs a thermoplastic polyurethane (TPU) film embedded with hafnium oxide nanoparticles (HfO2 NPs) to facilitate dynamic tracking of tumor growth and regression in real time. Significantly, the synthesized HfO2 NPs exhibit promising characteristics as effective sonosensitizers, holding the potential to efficiently eliminate cancer cells through ultrasound irradiation. The TPU-HfO2 film, acting as a dielectric elastomer (DE) strain sensor, undergoes proportional deformation in response to changes in the tumor volume, thereby influencing its electrical impedance. This distinctive behavior empowers the DE strain sensor to continuously and accurately monitor alterations in tumor volume, determining the optimal timing for initiating HfO2 NP treatment, optimizing dosages, and assessing treatment effectiveness. Seamless integration with a wireless system allows instant transmission of detected electrical impedances to a smartphone for real-time data processing and visualization, enabling immediate patient monitoring and timely intervention by remote medical staff. By combining the dynamic tumor monitoring capabilities of the TPU-HfO2 film with the sonosensitizer potential of HfO2 NPs, this approach propels cancer care into the realm of telemedicine, representing a significant advancement in patient treatment.


Assuntos
Nanopartículas , Neoplasias , Dispositivos Eletrônicos Vestíveis , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Ondas Ultrassônicas
2.
Int J Biol Macromol ; 224: 927-937, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36306906

RESUMO

Low-molecular-weight chitosan (LMWCS) damaged cell membranes in zebrafish showed its possibility to release reporter proteins for detection. In this study, we developed a simple fluorometric-based assay for the evaluation of clinical antiangiogenic drugs using LMWCS and Tg(fli1:EGFP) transgenic zebrafish, which expressed green-fluorescence protein (GFP) in the endothelial cells of blood vessel. In vitro stable and transiently transfected cell lines was released luciferase and green fluorescent protein (GFP) for intensity evaluation upon LMWCS fluorometric-based assay. In vivo Tg(fli1:EGFP) transgenic zebrafish was also released GFP from endothelial cells of blood vessels and show an increase of fluorescent intensity upon LMWCS fluorometric-based assay. Treatment with the clinical antiangiogenic drug sorafenib and analyzed by LMWCS fluorometric-based assay showed significantly reduction of angiogenesis. Furthermore, treatment with 2 µM sorafenib showed a significant reduction in angiogenesis of the intersegmental vein (ISV) and dorsal longitudinal anastomotic vessels (DLAV) in Tg(fli1:EGFP) transgenic zebrafish. Fluorescence intensity reduction from 2 µM sorafenib was used as a factor in the LMWCS fluorescence-based assay for relative antiangiogenic evaluation. Relative angiogenesis evaluation of the clinical drugs axitinib, cabozantinib, and regorafenib showed a significant reduction. Collectively, this study provided a simple, convenient, and rapid LMWCS fluorometric-based assay for evaluating angiogenic drugs using transgenic zebrafish.


Assuntos
Inibidores da Angiogênese , Quitosana , Animais , Peixe-Zebra/metabolismo , Células Endoteliais/metabolismo , Sorafenibe , Animais Geneticamente Modificados , Proteínas de Fluorescência Verde/metabolismo
3.
Int J Biol Macromol ; 221: 121-134, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36049568

RESUMO

Photothermal and photodynamic therapies (PTT/PDT) have been widely accepted as noninvasive therapeutic methods for cancer treatment. However, tumor hypoxia and insufficient delivery of photoactive compounds to cancer cells can reduce the efficacy of phototherapy. Herein, we first synthesized thiolated hyaluronic acid (THA) and then conjugated it with catalase (CAT) onto chlorin e6 (Ce6)-adsorbed small gold nanorods (Ce6@sAuNRs) with near-infrared (NIR)/visible light activated photothermal/photodynamic effects. The conjugation of THA and CAT on Ce6@sAuNRs resulted in a red-shift of the longitudinal LSPR absorption band of sAuNRs up to 1000 nm and maintained the excellent enzymatic activity of catalase. Modification of Ce6@sAuNRs with THA resulted in efficient internalization of the nanocomposite into MCF-7/ADR multidrug-resistant (MDR) breast cancer cells (CD44+), thereby significantly enhancing the intracellular accumulation of the photosensitizer Ce6. CAT endows Ce6@sAuNRs with self-supporting oxygen production, which enables them to efficiently generate singlet oxygen (1O2) under 660 nm laser irradiation and enhances the photodynamic effect against hypoxic breast cancer cells. The results highlight the prospect of this novel multi-functional nanoplatform integrating active biological macromolecules (THA and CAT) into photosensitizer/photothermal gold nanocomposites in overcoming the limitations of hypoxic MDR breast cancer cell treatment.


Assuntos
Neoplasias da Mama , Fotoquimioterapia , Porfirinas , Catalase , Ouro/farmacologia , Ácido Hialurônico/farmacologia , Oxigênio , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes , Porfirinas/farmacologia , Neoplasias da Mama/tratamento farmacológico , Humanos , Receptores de Hialuronatos , Nanotubos , Células MCF-7
4.
ACS Nano ; 16(8): 12403-12414, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35920682

RESUMO

The in situ transformation of low-toxicity precursors into a chemotherapeutic agent at a tumor site to enhance the efficacy of its treatment has long been an elusive goal. In this work, a zinc-based zeolitic imidazolate framework that incorporates pharmaceutically acceptable precursors is prepared as a nanoreactor (NR) system for the localized synthesis of an antitumor drug. The as-prepared NRs are administered intratumorally in a tumor-bearing mouse model and then irradiated with ultrasound (US) to activate the chemical synthesis. The US promotes the penetration of the administered NRs into the tumor tissue to cover the lesion entirely, although some NRs leak into the surrounding normal tissue. Nevertheless, only the tumor tissue, where the H2O2 concentration is high, is adequately exposed to the as-synthesized antitumor drug, which markedly impedes development of the tumor. No significant chemical synthesis is detected in the surrounding normal tissue, where the local H2O2 concentration is negligible and the US irradiation is not directly applied. The as-proposed tumor-specific in situ synthesis of therapeutic molecules induces hardly any significant in vivo toxicity and, thus, is potentially a potent biocompatible approach to precision chemotherapy.


Assuntos
Antineoplásicos , Neoplasias , Zeolitas , Camundongos , Animais , Portadores de Fármacos/química , Peróxido de Hidrogênio/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Zeolitas/química , Nanotecnologia
6.
Int J Biol Macromol ; 194: 384-394, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34822829

RESUMO

Many challenges, such as virus infection, extreme weather and long cultivation periods, during the development of fish larvae have been observed, especially in aquaculture. Gene delivery is a useful method to express functional genes to defend against these challengers. However, the methods for fish larvae are insufficient. In our earlier report, low-molecular-weight chitosan (LMWCS) showed a strong positive charge and may be useful for polyplex formulation. Herein, we present a simple self-assembly of LMWCS polyplexes (LMWCSrNPs) for gene delivery into zebrafish larvae. Different weight ratios of LMWCS/gamma-polyglutamic acid (γ-PGA)/plasmid DNA were analyzed by gel mobility assay. Delivery efficiency determined by green fluorescent protein (GFP) expression in zebrafish liver (ZFL) cells showed that delivery efficiency at a weight ratio of 20:8:1 was higher than others. Zeta potential and transmission electron microscopy (TEM) analysis showed that the round shape of the particle size varied. In our earlier reports, IRF9S2C could induce interferon-stimulated gene (ISG) expression to induce innate immunity in zebrafish and pufferfish. Further delivery of pcDNA3-IRF9S2C-HA plasmid DNA into ZFL cells and zebrafish larvae by LMWCSrNP successfully induced ISG expression. Collectively, LMWCSrNP could be a novel gene delivery system for zebrafish larvae and might be used to improve applications in aquaculture.


Assuntos
Quitosana/química , Portadores de Fármacos/química , Técnicas de Transferência de Genes , Ácidos Nucleicos/administração & dosagem , Ácido Poliglutâmico/análogos & derivados , Animais , Sobrevivência Celular , Células Cultivadas , Fenômenos Químicos , Portadores de Fármacos/síntese química , Expressão Gênica , Genes Reporter , Larva , Peso Molecular , Ácido Poliglutâmico/síntese química , Ácido Poliglutâmico/química , Análise Espectral , Peixe-Zebra
7.
Acta Biomater ; 134: 686-701, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34358695

RESUMO

Thrombolysis is a standard treatment for rapidly restoring blood flow. However, the application of urokinase-type plasminogen activator (Uk) in clinical therapy is limited due to its nonspecific distribution and inadequate therapeutic accumulation. Precise thrombus imaging and site-specific drug delivery can enhance the diagnostic and therapeutic efficacy for thrombosis. Accordingly, we developed a P-selectin-specific, photothermal theranostic nanocomposite for thrombus-targeted codelivery of Uk and indocyanine green (ICG, a contrast agent for near-infrared (NIR) fluorescence imaging). We evaluated its capabilities for thrombus imaging and enzyme/hyperthermia combined thrombolytic therapy. Mesoporous silica-coated gold nanorods (Si-AuNRs) were functionalized with an arginine-rich peptide to create an organic template for the adsorption of ICG and fucoidan (Fu), an algae-derived anticoagulant. Uk was loaded into the SiO2 pores of the Si-AuNRs through the formation of a Fu-Uk-ICG complex on the peptide-functionalized template. The Fu-Uk/ICG@SiAu NRs nanocomposite increased the photostability of ICG and improved its targeting/accumulation at blood clot sites with a strong NIR fluorescence intensity for precise thrombus imaging. Furthermore, ICG incorporated into the nanocomposite enhanced the photothermal effect of Si-AuNRs. Fu, as a P-selectin-targeting ligand, enabled the nanocomposite to target a thrombus site where platelets were activated. The nanocomposite enabled a faster release of Uk for rapid clearing of blood clots and a slower release of Fu for longer lasting prevention of thrombosis regeneration. The nanocomposite with multiple functions, including thrombus-targeting drug delivery, photothermal thrombolysis, and NIR fluorescence imaging, is thus an advanced theranostic platform for thrombolytic therapy with reduced hemorrhaging risk and enhanced imaging/thrombolysis efficiency. STATEMENT OF SIGNIFICANCE: Herein, for the first time, a P-selectin specific, photothermal theranostic nanocomposite for thrombus-targeted co-delivery of urokinase and NIR fluorescence contrast agent indocyanine green (ICG) was developed. We evaluated the potential of this theranostic nanocomposite for thrombus imaging and enzyme/hyperthermia combined thrombolytic therapy. The nanocomposite showed multiple functions including thrombus targeting and imaging, and photothermal thrombolysis. Besides, it allowed faster release of the thrombolytic urokinase for rapidly clearing blood clots and slower release of a brown algae-derived anticoagulant fucoidan (also acting as a P-selectin ligand) for prevention of thrombosis regeneration. The nanocomposite is thus a new and advanced theranostic platform for targeted thrombolytic therapy.


Assuntos
Nanocompostos , Nanopartículas , Trombose , Anticoagulantes/farmacologia , Linhagem Celular Tumoral , Meios de Contraste , Fibrinolíticos/farmacologia , Humanos , Verde de Indocianina , Fototerapia , Medicina de Precisão , Dióxido de Silício , Nanomedicina Teranóstica , Trombose/diagnóstico por imagem , Trombose/tratamento farmacológico
8.
Adv Mater ; 33(34): e2100701, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34270814

RESUMO

Most orally administered drugs fail to reach the intracerebral regions because of the intestinal epithelial barrier (IEB) and the blood-brain barrier (BBB), which are located between the gut and the brain. Herein, an oral prodrug delivery system that can overcome both the IEB and the BBB noninvasively is developed for treating gliomas. The prodrug is prepared by conjugating an anticancer drug on ß-glucans using a disulfide-containing linker. Following oral administration in glioma-bearing mice, the as-prepared prodrug can specifically target intestinal M cells, transpass the IEB, and be phagocytosed/hitchhiked by local macrophages (Mϕ). The Mϕ-hitchhiked prodrug is transported to the circulatory system via the lymphatic system, crossing the BBB. The tumor-overexpressed glutathione then cleaves the disulfide bond within the prodrug, releasing the active drug, improving its therapeutic efficacy. These findings reveal that the developed prodrug may serve as a gut-to-brain oral drug delivery platform for the well-targeted treatment of gliomas.


Assuntos
Administração Oral , Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Glioma/tratamento farmacológico , Intestinos/efeitos dos fármacos , Pró-Fármacos/química , Temozolomida/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Barreira Hematoencefálica/efeitos dos fármacos , Dissulfetos , Endocitose , Sistema Linfático , Macrófagos/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos , Transplante de Neoplasias , Temozolomida/farmacocinética , beta-Glucanas/química
9.
Int J Biol Macromol ; 166: 98-107, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33091478

RESUMO

Nanomaterial mediated cancer/tumor photo driven hyperthermia has obtained great awareness. Nevertheless, it is a challenge for improving the hyperthermic efficacy lacking resistance to stimulated thermal stress. We thus developed a bioinspired nano-platform utilizing inclusion complexation between photosensitive polypyrrole (Ppy) nanoparticles (NP) and fucoidan (FU). This FU-Ppy NP proved to be an excellent P-selectin-mediated, lung cancer-cell/tumor targeting delivery and specific accumulation, could augment cancer/tumor oxidative stress levels through producing cellular reactive oxygen species. Potent ROS/photothermal combinational therapeutic effects were exhibited by the bioinspired FU-Ppy NP through a selective P-selectin cancer/tumor targeting aptitude for the lung cancer cells/tumor compared with other nano-formulations. The usage of FU-Ppy NP also involves the potential mechanism of suppressing the biological expression of tumor vascular endothelial growth factor (VEGF). This FU biological macromolecule-amplified photothermally therapeutic nano-platform has promising potential for future medical translation in eradicating numerous tumors.


Assuntos
Neoplasias Pulmonares/terapia , Nanopartículas/química , Terapia Fototérmica/métodos , Polímeros/química , Polissacarídeos/química , Pirróis/química , Espécies Reativas de Oxigênio/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Luz , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Nanopartículas/efeitos da radiação , Nanopartículas/uso terapêutico , Selectina-P/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Int J Mol Sci ; 21(21)2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158139

RESUMO

Galectin-3 (Gal-3) is a 26-kDa lectin that regulates many aspects of inflammatory cell behavior. We assessed the hypothesis that increased levels of Gal-3 contribute to abdominal aortic aneurysm (AAA) progression by enhancing monocyte chemoattraction through macrophage activation. We analyzed the plasma levels of Gal-3 in 76 patients with AAA (AAA group) and 97 controls (CTL group) as well as in angiotensin II (Ang-II)-infused ApoE knockout mice. Additionally, conditioned media (CM) were used to polarize THP-1 monocyte to M1 macrophages with or without Gal-3 inhibition through small interfering RNA targeted deletion to investigate whether Gal-3 inhibition could attenuate macrophage-induced inflammation and smooth muscle cell (SMC) apoptosis. Our results showed a markedly increased expression of Gal-3 in the plasma and aorta in the AAA patients and experimental mice compared with the CTL group. An in vitro study demonstrated that the M1 cells exhibited increased Gal-3 expression. Gal-3 inhibition markedly decreased the quantity of macrophage-induced inflammatory regulators, including IL-8, TNF-α, and IL-1ß, as well as messenger RNA expression and MMP-9 activity. Moreover, Gal-3-deficient CM weakened SMC apoptosis through Fas activation. These findings prove that Gal-3 may contribute to AAA progression by the activation of inflammatory macrophages, thereby promoting SMC apoptosis.


Assuntos
Aneurisma da Aorta Abdominal/patologia , Apoptose/fisiologia , Proteínas Sanguíneas/fisiologia , Galectinas/fisiologia , Ativação de Macrófagos/fisiologia , Miócitos de Músculo Liso/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/sangue , Aneurisma da Aorta Abdominal/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Feminino , Galectinas/sangue , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/patologia
11.
Mater Sci Eng C Mater Biol Appl ; 114: 111064, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32994013

RESUMO

P-selectin overexpressed on activated endothelial cells and platelets is a new target for treatment of cancers and cardiovascular diseases such as atherosclerosis and thrombosis. In this study, depolymerized low molecular weight fucoidan (LMWF8775) and a thermolysin-hydrolyzed protamine peptide (TPP1880) were prepared. TPP1880 and LMWF8775 were able to form self-assembled complex nanoparticles (CNPs). The formation of TPP1880/LMWF8775 CNPs was characterized by Fourier-transform infrared spectra, circular dichroism spectra and isothermal titration calorimetry. The CNPs selectively targeted PMA-stimulated, inflamed endothelial cells (HUVECs) with high expression of P-selectin. Gd-DTPA MRI contrast agent was successfully loaded in the CNPs with better T1 relaxivity and selectively accumulated in the activated HUVECs with increased MRI intensity and reduced cytotoxicity as compared to free Gd-DTPA. Our results suggest that the TPP1880/LMWF8775 CNPs may have potential in future for early diagnosis of cardiovascular diseases and cancers in which the endothelium is inflamed or activated.


Assuntos
Gadolínio DTPA , Nanopartículas , Meios de Contraste , Células Endoteliais , Endotélio , Imageamento por Ressonância Magnética , Peptídeos , Polissacarídeos
12.
Biomaterials ; 257: 120227, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32738653

RESUMO

Multifunctional nanoplatforms combined with photodynamic therapy (PDT) and anticancer drugs have shown great promising in cancer therapy. However, their efficacy is limited by the low specificity, low oxygen levels, and a tolerant tumor immune microenvironment. Herein, we developed a biocompatible theranostic nanoplatform (FM@VP) based on co-assembly of a nanocomplex formed by a functional polysaccharide fucoidan and a bioreducible polyamidoamine (PAMAM) dendrimer, a photosensitizer verteporfin (VP), and MnO2 nanoparticles (a tumor microenvironment responsive oxygen evolving nanomaterial) into a multifunctional nanoparticle cluster. The dendrimer-fucoidan polyionic nanocomplex (DFPN) specifically targeted P-selectin-overexpressed triple-negative breast cancer (TNBC) and the tumor-associated vasculature, and was sensitive to glutathione (GSH) in tumor. More importantly, this FM@VP nanocomplex simultaneously overcame tumor hypoxia, suppressed oncogenic signaling, and attenuated tumor-mediated immunosuppression, resulting in improving therapeutic efficacy of PDT while enhancing antitumor immunity and anti-metastasis. This discovery provides a powerful strategy for synergetic cancer targeting/photodynamic/immunotherapy and could serve as a safe clinical translational approach.


Assuntos
Nanopartículas , Fotoquimioterapia , Linhagem Celular Tumoral , Humanos , Hipóxia/tratamento farmacológico , Compostos de Manganês , Óxidos , Fármacos Fotossensibilizantes/uso terapêutico , Polissacarídeos
13.
Biomaterials ; 255: 120157, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32535305

RESUMO

The therapeutic outcome of pancreatic cancer remains unsatisfactory, despite many attempts to improve it. To address this challenge, an oral drug delivery system that spontaneously initiates an effervescent reaction to form gas-bubble carriers is proposed. These carriers concurrently deliver lipophilic paclitaxel (PTX) and hydrophilic gemcitabine (GEM) in the small intestine. The bursting of the bubbles promotes the intestinal absorption of the drugs. The antitumor efficacy of this proposed oral drug delivery system is evaluated in rats with experimentally created orthotopic pancreatic tumors. The combined administration of equivalent amounts of PTX and GEM via the intravenous (i.v.) route, which is clinically used for treating pancreatic cancer, serves as a control. Following oral administration, the lipophilic PTX is initially absorbed through the intestinal lymphatic system and then enters systemic circulation, whereas the hydrophilic GEM is directly taken up into the blood circulation, ultimately accumulating in the tumorous pancreatic tissues. A pharmacokinetic study reveals that the orally delivered formulation has none of the toxic side-effects that are associated with the i.v. injected formulation; changes the pharmacokinetic profiles of the drugs; and increases the bioavailability of PTX. The oral formulation has a greater impact than the i.v. formulation on tumor-specific stromal depletion, resulting in greater inhibition of tumor growth with no evidence of metastatic spread. As well as enhancing the therapeutic efficacy, this unique approach of oral chemotherapy has potential for use on outpatients, greatly improving their quality of life.


Assuntos
Neoplasias Pancreáticas , Qualidade de Vida , Administração Oral , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/uso terapêutico , Sistemas de Liberação de Medicamentos , Interações Hidrofóbicas e Hidrofílicas , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Ratos
14.
Carbohydr Polym ; 240: 116164, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32475540

RESUMO

Chitosan is suggested as no or low toxicity and biocompatible biomaterial. Digestion of chitosan to reduce molecular weight and formulate nanoparticle was generally used to improve efficiency for DNA or protein delivery. However, the toxicity of low-molecular-weight chitosan (LMWCS) towards freshwater fishes has not been well evaluated. Here, we reported the toxic mechanism of LMWCS using zebrafish (Danio rerio) liver (ZFL) cell line, zebrafish larvae, and adult fish. LMWCS rapidly induced cytotoxicity of ZFL cells and death of zebrafish. Cell membrane damaged by LMWCS reduced cell viability. Damaged membrane of epithelial cell in zebrafish larvae induced breakage of the yolk. Adult fish exhibited hypoxia before death due to multiple damages induced by LMWCS. Although the toxicity of LMWCS was revealed in zebrafish model, the toxicity was only present in pH < 7 and easy be neutralized by other negative ions. Collectively, these data improved a new understanding of LMWCS properties.


Assuntos
Materiais Biocompatíveis/toxicidade , Quitosana/toxicidade , Larva/efeitos dos fármacos , Fígado/efeitos dos fármacos , Peixe-Zebra/metabolismo , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Quitosana/análogos & derivados , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Peso Molecular , Testes de Toxicidade
15.
Int J Biol Macromol ; 160: 558-570, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32464213

RESUMO

The polysaccharides from Ophiopogon japonicus (OJPs) were known to have protective effects against diabetes, and cardiovascular and chronic inflammatory diseases. However, OJPs were poorly absorbed after oral administration, resulting in limited efficacy because of the low bioavailability. In this study, OJPs extracted and fractionated from Ophiopogon japonicus were used to prepare OJPs/chitosan (CS)/whey protein (WP) co-assembled nanoparticles. The OJPs/CS/WP nanoparticles showed high biocompatibility and inhibited the cytotoxicity of RAW264.7 cells induced by nickel. With the assistance of CS and WP, the anti-inflammatory and antioxidant activities of OJPs were enhanced because the nanoparticles improved OJPs uptake by RAW264.7 macrophage cells as evidenced by efficient scavenging of DPPH and ABTS free radicals and effective inhibition of NO production and the gene expressions of iNOS, COX2, TNF-α, CCL2, and CXCL2 inflammatory signals. Determining the transepithelial electrical resistance and paracellular permeability of Caco-2 monolayer/macrophage co-cultured system suggested that the OJPs-loaded nanoparticles effectively protected the intestinal epithelial barrier integrity against the damage caused by LPS-stimulated macrophage inflammation and attenuated the defects of intestinal epithelial TJ barrier and permeability. These findings suggest that the OJPs/CS/WP nanoparticles may be potential carriers for oral delivery of OJPs to treat intestinal barrier defects, such as inflammatory bowel disease (IBD).


Assuntos
Quitosana/química , Células Epiteliais/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Nanopartículas/química , Ophiopogon/química , Polissacarídeos/administração & dosagem , Junções Íntimas/efeitos dos fármacos , Proteínas do Soro do Leite/química , Administração Oral , Animais , Disponibilidade Biológica , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Mucosa Intestinal/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Permeabilidade/efeitos dos fármacos , Polissacarídeos/química , Células RAW 264.7
16.
Int J Biol Macromol ; 149: 600-608, 2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32004612

RESUMO

Fucoidan is a fucose-rich polysaccharide that has gained attention for its various anticancer properties. However, the effect and underlying mechanism of fucoidan on triple-negative breast cancer (TNBC) are still unknown. Herein, we investigated the anticancer potential of fucoidan from Laminaria japonica. We found that fucoidan showed modest antiproliferative activity against TNBC cells, while it effectively reduced migratory and invasive capacities. Mechanistically, fucoidan suppressed activation of MAPK and PI3K followed by inhibition of AP-1 and NF-κB signaling in TNBC. Additionally, fucoidan downregulated expressions of proangiogenic factors in TNBC cells, and fucoidan blocked tumor-elicited tube formation by human umbilical vascular endothelial cells (HUVECs). We also observed that fucoidan blocked tumor adhesion and invasion towards HUVECs. Surprisingly, fucoidan robustly suppressed tube formation on HUVECs. Moreover, fucoidan inhibited in vivo angiogenesis and micrometastasis in a transgenic zebrafish model. Together, L. japonica fucoidan exhibits potent antitumor effects by its attenuation of invasiveness and proangiogenesis in TNBC.


Assuntos
Laminaria/química , Neovascularização Patológica/tratamento farmacológico , Polissacarídeos/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , NF-kappa B/genética , Micrometástase de Neoplasia , Neovascularização Patológica/patologia , Polissacarídeos/química , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/genética , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Acta Biomater ; 96: 468-479, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31260820

RESUMO

Fibrinolytic treatments for venous or arterial thrombotic syndromes using systemic administration of thrombolytics, such as streptokinase, can induce life-threatening bleeding complications. In this study, we offer the first proof of concept for a targeted photothermal fibrin clot prevention and reduction technology using macrophages loaded with polypyrrole-polyethylenimine nanocomplexes (Ppy-PEI NCs) and subjected to near-infrared radiation (NIR). We first show that the developed Ppy-PEI NCs could be taken up by defensive macrophages in vitro through endocytosis. The Ppy-PEI NCs generated local hyperthermia upon NIR treatment, which appeared to produce reactive oxygen species in Ppy-PEI NC-loaded macrophages. Preliminary evidence of efficacy as an antithrombotic tool is provided, in vitro, using fibrinogen-converted fibrin clots, and in vivo, in a rat femoral vascular thrombosis model generated by exposure to ferric chloride substance. The in vivo biocompatibility, photothermal behavior, biodistribution, and histological observation of cellular interactions with the Ppy-PEI NCs in the rat model provide rationale in support of further preclinical studies. This Ppy-PEI NC/NIR-based method, which uses a unique macrophage-guided targeting approach to prevent and lyse fibrin clots, may potentially overcome some of the disadvantages of current thrombolytic treatments. STATEMENT OF SIGNIFICANCE: Fibrinolytic treatments for venous or arterial thrombotic syndromes using systemic administration of thrombolytics, such as streptokinase, can induce life-threatening bleeding complications. In this study, we offer the first proof of concept for a targeted photothermal fibrin clot reduction technology using macrophages loaded with polypyrrole-polyethylenimine nanocomplexes (Ppy-PEI NCs) and subjected to near-infrared radiation (NIR). We first show that the developed Ppy-PEI NCs can be taken up by defensive macrophages in vitro through endocytosis. The Ppy-PEI NCs generated local hyperthermia upon NIR treatment, which appeared to produce reactive oxygen species in Ppy-PEI NC-loaded macrophages. Preliminary evidence of efficacy as an antithrombotic tool is provided, in vitro, using fibrinogen-converted fibrin clots, and in vivo, in a rat femoral vascular thrombosis model generated by exposure to ferric chloride substance. The in vivo biocompatibility, photothermal behavior, biodistribution, and histological observation of cellular interactions with the Ppy-PEI NCs in the rat model provide rationale in support of further preclinical studies. This Ppy-PEI NC/NIR-based method, which uses a unique macrophage-guided targeting approach to disintegrate fibrin clots, may potentially overcome some of the disadvantages of current thrombolytic treatments.


Assuntos
Materiais Biomiméticos/química , Fibrina/metabolismo , Hipertermia Induzida , Macrófagos/citologia , Nanopartículas/química , Polietilenoimina/farmacologia , Polímeros/farmacologia , Pirróis/farmacologia , Trombose/prevenção & controle , Animais , Endocitose , Humanos , Raios Infravermelhos , Camundongos , Camundongos Endogâmicos ICR , Nanopartículas/ultraestrutura , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Temperatura , Distribuição Tecidual
18.
Carbohydr Polym ; 193: 163-172, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-29773368

RESUMO

Self-assembled nanoparticles (NPs) from arginine-modified chitosan (CS-N-Arg) and thiolated fucoidan (THL-fucoidan) were synthesized to enhance the transport of dextran and curcumin across intestinal epithelial cell layer. CS-N-Arg/THL-fucoidan NPs exhibited a pH-sensitive assembly-disassembly and drug release property. Evaluations of the NPs in enhancing the transport of a hydrophilic macromolecule (FITC-dextran) and a hydrophobic drug (curcumin) were investigated in Caco-2 cell monolayers. The cationic CS-N-Arg in the NPs induced disruption of intestinal epithelial tight junctions as indicated by the decrease of transepithelial electrical resistance (TEER). Permeation studies revealed that the NPs enhanced the paracellular permeation of macromolecular dextran through the monolayer barrier. In addition, the multifunctional NPs increased the permeability of rhodamine 123 because the thiomer THL-fucoidan in the NPs inhibited P-glycoprotein. Cellular uptake and permeability of curcumin encapsulated in the NPs were improved due to increasing their water solubility and stability.


Assuntos
Quitosana/química , Curcumina/administração & dosagem , Dextranos/administração & dosagem , Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Nanopartículas/química , Polissacarídeos/química , Compostos de Sulfidrila/química , Administração Oral , Arginina/administração & dosagem , Arginina/química , Sobrevivência Celular/efeitos dos fármacos , Quitosana/administração & dosagem , Curcumina/farmacologia , Dextranos/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Intestinos/efeitos dos fármacos , Estrutura Molecular , Tamanho da Partícula , Polissacarídeos/administração & dosagem , Relação Estrutura-Atividade , Compostos de Sulfidrila/administração & dosagem , Propriedades de Superfície
19.
ACS Biomater Sci Eng ; 4(8): 2847-2859, 2018 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-33435008

RESUMO

The clinical treatment of gastric cancer is hampered by the development of anticancer drug resistance as well as the unfavorable pharmacokinetics, nontarget toxicity, and inadequate intratumoral accumulation of current chemotherapies. The polyphenol epigallocatechin gallate in combination with doxorubicin exhibits synergistic inhibition P-glycoprotein efflux pump activity and cancer cell growth. This study evaluated a potential activated nanoparticle delivery system comprising a hyaluronic acid complex with polyethylene glycol-conjugated gelatin containing encapsulated epigallocatechin gallate and low-dose doxorubicin, which may facilitate targeted drug administration to gastric cancer cells. We confirmed successful delivery of bioactive combination drugs and internalization into gastric cancer cells through CD44 ligand recognition and ensuing inhibition of cell proliferation via caspase-induced apoptosis and G2/M phase cell cycle arrest. Furthermore, the targeted nanoparticles significantly suppressed gastric tumor activity and reduced both tumor and heart tissue inflammatory reaction in vivo compared to systemic combination treatment.

20.
Carbohydr Polym ; 180: 286-296, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-29103507

RESUMO

Bacterial cellulose (BC) is a biopolymer composed of nanofibers which has excellent film-forming ability. However, BC do not have antibacterial or antioxidant activity, thus limiting the applicability of BC for food and biomedical applications. In this study, flavonoid silymarin (SMN) and zein were assembled into spherical SMN-Zein nanoparticles that could be effectively adsorbed onto BC nanofibers. SMN-Zein nanoparticles greatly changed the wettability and swelling property of BC films due to the formation of nanoparticles/nanofibers nanocomposites. SMN-Zein nanoparticles enhanced the release of sparingly soluble silymarin from the nanocomposite films. The active films showed more effective antioxidant and antibacterial activities as compared with pure BC films and thus were able to protect salmon muscle from deterioration and lipid oxidation. These findings suggest that the nanoparticle/nanofiber composites may offer a suitable platform for modification of BC films with improved drug release properties and biological activities.


Assuntos
Antibacterianos/química , Antioxidantes/química , Celulose/análogos & derivados , Conservação de Alimentos/métodos , Nanocompostos/química , Silimarina/química , Zeína/química , Antibacterianos/farmacologia , Antioxidantes/farmacologia , Liberação Controlada de Fármacos , Produtos Pesqueiros , Gluconacetobacter xylinus/química , Nanofibras/química , Nanopartículas/química , Polissacarídeos Bacterianos/química , Pseudomonas/efeitos dos fármacos , Silimarina/farmacologia , Staphylococcus/efeitos dos fármacos , Molhabilidade , Zeína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA