Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Healthc Mater ; : e2304675, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38688026

RESUMO

The mitochondrial enzyme arginase-2 (Arg-2) is implicated in the pathophysiology of contrast-induced acute kidney injury (CI-AKI). Therefore, Arg-2 represents a candid target for CI-AKI prevention. Here, layer-by-layer (LbL) assembled renal-targeting polymeric nanoparticles are developed to efficiently deliver small interfering RNA (siRNA), knockdown Arg-2 expression in renal tubules, and prevention of CI-AKI is evaluated. First, near-infrared dye-loaded poly(lactic-co-glycolic acid) (PLGA) anionic cores are electrostatically coated with cationic chitosan (CS) to facilitate the adsorption and stabilization of Arg-2 siRNA. Next, nanoparticles are coated with anionic hyaluronan (HA) to provide protection against siRNA leakage and shielding against early clearance. Sequential electrostatic layering of CS and HA improves loading capacity of Arg-2 siRNA and yields LbL-assembled nanoparticles. Renal targeting and accumulation is enhanced by modifying the outermost layer of HA with a kidney targeting peptide (HA-KTP). The resultant kidney-targeting and siRNA loaded nanoparticles (PLGA/CS/HA-KTP siRNA) exhibit proprietary accumulation in kidneys and proximal tubular cells at 24 h post-tail vein injection. In iohexol-induced in vitro and in vivo CI-AKI models, PLGA/CS/HA-KTP siRNA delivery alleviates oxidative and nitrification stress, and rescues mitochondrial dysfunction while reducing apoptosis, thereby demonstrating a robust and satisfactory therapeutic effect. Thus, PLGA/CS/HA-KTP siRNA nanoparticles offer a promising candidate therapy to protect against CI-AKI.

2.
J Nanobiotechnology ; 19(1): 451, 2021 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-34961540

RESUMO

BACKGROUND: Hypoxia is a major contributor to global kidney diseases. Targeting hypoxia is a promising therapeutic option against both acute kidney injury and chronic kidney disease; however, an effective strategy that can achieve simultaneous targeted kidney hypoxia imaging and therapy has yet to be established. Herein, we fabricated a unique nano-sized hypoxia-sensitive coassembly (Pc/C5A@EVs) via molecular recognition and self-assembly, which is composed of the macrocyclic amphiphile C5A, the commercial dye sulfonated aluminum phthalocyanine (Pc) and mesenchymal stem cell-excreted extracellular vesicles (MSC-EVs). RESULTS: In murine models of unilateral or bilateral ischemia/reperfusion injury, MSC-EVs protected the Pc/C5A complex from immune metabolism, prolonged the circulation time of the complex, and specifically led Pc/C5A to hypoxic kidneys via surface integrin receptor α4ß1 and αLß2, where Pc/C5A released the near-infrared fluorescence of Pc and achieved enhanced hypoxia-sensitive imaging. Meanwhile, the coassembly significantly recovered kidney function by attenuating cell apoptosis, inhibiting the progression of renal fibrosis and reducing tubulointerstitial inflammation. Mechanistically, the Pc/C5A coassembly induced M1-to-M2 macrophage transition by inhibiting the HIF-1α expression in hypoxic renal tubular epithelial cells (TECs) and downstream NF-κB signaling pathway to exert their regenerative effects. CONCLUSION: This synergetic nanoscale coassembly with great translational potential provides a novel strategy for precise kidney hypoxia diagnosis and efficient kidney injury treatment. Furthermore, our strategy of coassembling exogenous macrocyclic receptors with endogenous cell-derived membranous structures may offer a functional platform to address multiple clinical needs.


Assuntos
Injúria Renal Aguda/diagnóstico por imagem , Injúria Renal Aguda/tratamento farmacológico , Hipóxia Celular/efeitos dos fármacos , Vesículas Extracelulares/química , Compostos Macrocíclicos/química , Tensoativos/química , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Calixarenos/química , Calixarenos/metabolismo , Calixarenos/farmacologia , Calixarenos/uso terapêutico , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Indóis/química , Indóis/metabolismo , Indóis/farmacologia , Indóis/uso terapêutico , Inflamação , Integrinas/metabolismo , Compostos Macrocíclicos/metabolismo , Compostos Macrocíclicos/farmacologia , Compostos Macrocíclicos/uso terapêutico , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Compostos Organometálicos/farmacologia , Compostos Organometálicos/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Tensoativos/metabolismo , Tensoativos/farmacologia , Tensoativos/uso terapêutico
3.
Zygote ; : 1-10, 2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31933449

RESUMO

Studies have indicated that psychological stress impairs human fertility and that various stressors can induce apoptosis of testicular cells. However, the mechanisms by which psychological stress on males reduces semen quality and stressors induce apoptosis in testicular cells are largely unclear. Using a psychological (restraint) stress mouse model, we tested whether male psychological stress triggers apoptosis of spermatozoa and spermatogenic cells through activating tumour necrosis factor (TNF)-α signalling. Wild-type or TNF-α-/- male mice were restrained for 48 h before examination for apoptosis and expression of TNF-α and TNF receptor 1 (TNFR1) in spermatozoa, epididymis, seminiferous tubules and spermatogenic cells. The results showed that male restraint significantly decreased fertilization rate and mitochondrial membrane potential, while increasing levels of malondialdehyde, active caspase-3, TNF-α and TNFR1 in spermatozoa. Male restraint also increased apoptosis and expression of TNF-α and TNFR1 in caudae epididymides, seminiferous tubules and spermatogenic cells. Sperm quality was also significantly impaired when spermatozoa were recovered 35 days after male restraint. The restraint-induced damage to spermatozoa, epididymis and seminiferous tubules was significantly ameliorated in TNF-α-/- mice. Furthermore, incubation with soluble TNF-α significantly reduced sperm motility and fertilizing potential. Taken together, the results demonstrated that male psychological stress induces apoptosis in spermatozoa and spermatogenic cells through activating the TNF-α system and that the stress-induced apoptosis in spermatogenic cells can be translated into impaired quality in future spermatozoa.

4.
ACS Appl Bio Mater ; 2(11): 5053-5061, 2019 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-35021503

RESUMO

This study evaluated the feasibility of microneedle (MN) technology as an alternative approach for pertussis toxin (PT) vaccine delivery. The characterization of PT vaccine MNs ensured sufficient mechanical property to deliver antigens to intradermal tissues of mice for intensive stimulations at the targeting of immune cells. The histological examination suggested that PT vaccine MNs successfully embedded into the mouse dermis to a depth of approximately 330 µm. The in vivo fluorescence imaging showed the sustained and longer release of antigens through MN patches compared to a direct subcutaneous (s.c.) injection, thus resulting in the quick generation of a high level of PT-specific IgG after the first immunization. Moreover, similar IgG levels could be elicited by these two delivery strategies, while less PT antigens were needed using MN patches. The cytokines studies illustrated that PT vaccine MNs could elicit immune responses through multiple pathways, especially of Th1 and Th17 pathways. By integrating the above results, we concluded that the PT MN transdermal drug delivery system could improve antigen utilization and induce rapid and robust PT-specific immune responses, which possessed the superiority over the traditional s.c. injection.

5.
Int J Nanomedicine ; 12: 6721-6733, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28932116

RESUMO

A 3-D scaffold that simulates the microenvironment in vivo for regenerating cartilage is ideal. In this study, we combined silk fibroin and decellularized cartilage extracellular matrix by temperature gradient-guided thermal-induced phase separation to produce composite scaffolds (S/D). Resulting scaffolds had remarkable mechanical properties and biomimeticstructure, for a suitable substrate for attachment and proliferation of adipose-derived stem cells (ADSCs). Moreover, transforming growth factor ß3 (TGF-ß3) loaded on scaffolds showed a controlled release profile and enhanced the chondrogenic differentiation of ADSCs during the 28-day culture. The S/D scaffold itself can provide a sustained release system without the introduction of other controlled release media, which has potential for commercial and clinical applications. The results of toluidine blue, Safranin O, and immunohistochemical staining and analysis of collagen II expression showed maintenance of a chondrogenic phenotype in all scaffolds after 28-day culture. The most obvious phenomenon was with the addition of TGF-ß3. S/D composite scaffolds with sequential delivery of TGF-ß3 may mimic the regenerative microenvironment to enhance the chondrogenic differentiation of ADSCs in vitro.


Assuntos
Matriz Extracelular/química , Fibroínas/química , Células-Tronco/citologia , Alicerces Teciduais/química , Fator de Crescimento Transformador beta3/administração & dosagem , Tecido Adiposo/citologia , Animais , Materiais Biomiméticos , Cartilagem Articular/citologia , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Colágeno/metabolismo , Coelhos , Células-Tronco/efeitos dos fármacos , Suínos , Fator de Crescimento Transformador beta3/farmacologia
6.
Biomaterials ; 113: 191-202, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27816821

RESUMO

Whole tumor cell lysates (TCL) have been implemented as tumor antigens for cancer vaccine development, although clinical outcomes of TCL-based antitumor immunotherapy remain unsatisfactory. In order to improve the efficacy of TCL-based vaccines, biomaterials have been employed to enhance antigen delivery and presentation. Here, we have developed chitosan nanoparticles (CTS NPs) with surface mannose (Man) moieties for specific dendritic cells (DCs) targeting (Man-CTS NPs). The Man-CTS NPs were then loaded with TCL generated from B16 melanoma cells (Man-CTS-TCL NPs) for in vitro and in vivo assessment. Potency of the Man-CTS-TCL NPs as cancer vaccine was also assessed in vivo by immunization of mice with Man-CTS-TCL NPs followed by re-challenge with B16 melanoma cell inoculation. We have shown here that Man-CTS-TCL NPs promote bone marrow-derived dendritic cells (BMDCs) maturation and antigen presentation in vitro. In vivo evaluation further demonstrated that the Man-CTS-TCL NPs were readily taken up by endogenous DCs within the draining lymph node (DLN) following subcutaneous administration accompanied by increasing in serum IFN-γ and IL-4 levels. Tumor growth was also significantly delayed in mice primed with Man-CTS-TCL NPs vaccine, attributable at least in part to cytotoxic T lymphocytes response. Moreover, Man-CTS-TCL NPs vaccine also exhibited therapeutic effects in mice with melanoma. Thus, we report here the Man-CTS-TCL NPs as effective anti-tumor vaccine for cancer immunotherapy.


Assuntos
Antígenos de Neoplasias/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Quitosana/química , Células Dendríticas/imunologia , Portadores de Fármacos/química , Melanoma Experimental/terapia , Nanopartículas/química , Animais , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Feminino , Imunoterapia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
7.
Oncotarget ; 7(15): 20934-44, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26988913

RESUMO

Cancer development and progression is linked to tumor-associated macrophages (TAMs). Distinct TAMs subsets perform either protective or pathogenic effects in cancer. A protective role in carcinogenesis has been described for M1 macrophages, which activate antitumor mechanisms. By comparison, TAMs isolated from solid and metastatic tumors have a suppressive M2-like phenotype, which could support multiple aspects of tumor progression. Currently, it has not been clearly understood how macrophages in tumor-associated stroma could be hijacked to support tumor growth. Mesenchymal stem cells (MSCs) actively interact with components of the innate immune system and display both anti-inflammatory and pro-inflammatory effects. Here, we tested whether MSCs could favor the tumor to escape from immunologic surveillance in the presence of M1 macrophages. We found that MSCs educated by M1 condition medium (cMSCs) possessed a greatly enhanced ability in promoting tumor growth in vivo. Examination of cytokines/chemokines showed that the cMSCs acquired a regulatory profile, which expressed high levels of iNOS and MCP1. Consistent with an elevated MCP1 expression in cMSCs, the tumor-promoting effect of the cMSCs depended on MCP1 mediated macrophage recruitment to tumor sites. Furthermore, IL-6 secreted by the cMSCs could polarize infiltrated TAMs into M2-like macrophages. Therefore, when macrophages changed into M1 pro-inflammation type in tumor microenvironment, the MSCs would act as poor sensors and switchers to accelerate tumor growth.


Assuntos
Neoplasias da Mama/imunologia , Carcinoma Hepatocelular/imunologia , Transformação Celular Neoplásica/imunologia , Glioblastoma/imunologia , Neoplasias Hepáticas/imunologia , Macrófagos/imunologia , Células-Tronco Mesenquimais/imunologia , Animais , Apoptose , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Quimiocinas/metabolismo , Meios de Cultivo Condicionados , Citocinas/metabolismo , Feminino , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Terapia de Imunossupressão , Interleucina-6/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Tumorais Cultivadas , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 6: 20764, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26857840

RESUMO

Although there are many reports on the effect of glucose metabolism on oocyte nuclear maturation, there are few studies on its effect on ooplasmic maturation. By manipulating glucose metabolism pathways using a maturation medium that could support oocyte nuclear maturation but only a limited blastocyst formation without glucose, this study determined effects of glucose metabolism pathways on ooplasmic maturation. During maturation of cumulus-oocyte-complexes (COCs) with glucose, the presence of PPP inhibitor, DHEA or glycolysis inhibitor, iodoacetate significantly decreased blastocyst rates, intraoocyte glutathione and ATP. While blastocyst rates, GSH/GSSG ratio and NADPH were higher, ROS was lower significantly in COCs matured with iodoacetate than with DHEA. Fructose-6-phosphate overcame the inhibitory effect of DHEA on PPP. During maturation of COCs with pyruvate, electron transport inhibitor, rotenone or monocarboxylate transfer inhibitor, 4-CIN significantly decreased blastocyst rates. Cumulus-denuded oocytes had a limited capacity to use glucose or lactate, but they could use pyruvate to support maturation. In conclusion, whereas glycolysis promoted ooplasmic maturation mainly by supplying energy, PPP facilitated ooplasmic maturation to a greater extent by both reducing oxidative stress and supplying energy through providing fructose-6-phosphate for glycolysis. Pyruvate was transferred by monocarboxylate transporters and utilized through mitochondrial electron transport to sustain ooplasmic maturation.


Assuntos
Células do Cúmulo/metabolismo , Citoplasma/metabolismo , Glucose/metabolismo , Oócitos/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Células do Cúmulo/citologia , Feminino , Camundongos , Oócitos/citologia
9.
Chin J Integr Med ; 19(8): 573-81, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22903442

RESUMO

OBJECTIVE: To investigate whether electroacupuncture (EA) can promote cell survival and enhance heart function of mesenchymal stem cells (MSCs) therapy. METHODS: MSCs were isolated from bone marrow and expanded in Minimum Essential Medium Alpha (α-MEM). MI was induced in 72 Sprague-Dawley (S-D) rats by ligation of the left anterior descending coronary artery (LAD) for 30 min and reperfusion. MI rats randomly received injection of 1×10(6) DiI-labeled MSCs alone (n =24, MSC group), or plus electroacupuncture (EA) at Neiguan (PC6, n=24, EA+MSC group), or saline (n =24, saline group). EA treatment was performed for 4 days. Another 24 rats were subjected to chest-open surgery without LAD occlusion and treatment (sham group). Three time points, 4, 14 and 28 days (n =8 for each group) were included in this study. The survival of transplanted MSCs and the protective gene expression were analyzed by reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot at day 4 and 14. Left ventricular remodeling, cardiac function, infarction area, fibrosis and capillary density were analyzed at day 28. RESULTS: EA can enhance MSC survival (2.6-fold up) at day 4. Big capillary density was 53% higher in EA+MSC treated group than MSC alone group. Furthermore, the rats treated by EA reduced the fibrosis and had 36% smaller infarct size comparing to MSC alone. EA also attenuated left ventricular remodeling and enhanced the functional recovery of infarcted hearts at week 4. CONCLUSION: EA at Neiguan acupoint can promote the stem cell survival and improve ischemic heart function. EA could become a useful approach in stem cell therapy for ischemia heart diseases.


Assuntos
Eletroacupuntura , Sobrevivência de Enxerto/fisiologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Isquemia Miocárdica/terapia , Animais , Apoptose/fisiologia , Sobrevivência Celular , Células Cultivadas , Terapia Combinada/métodos , Feminino , Coração/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Ratos , Ratos Sprague-Dawley , Remodelação Ventricular/fisiologia
10.
J Biomed Mater Res A ; 99(4): 684-9, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21913319

RESUMO

A novel folate fluorescent nanoconjugate was synthesized and used for detection of cancer cells overexpressing the folate receptor (FR). The folate conjugate (PCMS-NA-FA) was synthesized by conjugating folic acid (FA) and 4-ethylnyl-N-ethyl-1, 8-naphthalimide (NA) to the polychloromethylstyrene (PCMS) functionalized with azido group (PCMS-N3) through click reaction. The obtained conjugate had clear structure and could form PCMS-NA-FA nanoparticles with particle size around 86 nm in aqueous solution. Ability of PCMS-NA-FA targeting to cancerous cells was investigated by comparing the uptake of the nanoparticles by human adenocarcinoma HeLa cells and by non-FR expressing human lung carcinoma A549 cells. Specificity of the PCMS-NA-FA nanoparticles targeting on FRs was verified with cellular uptake inhibition assay, in which HeLa cells were incubated with both nanoconjugate and free FA. In addition, the specificity was also confirmed by the collocation of the immunofluorescence staining of anti-FR and the cellular uptake of the PCMS-NA-FA nanoparticles. Furthermore, the organ distribution of this folate nanoconjugate was studied on HeLa cell-bearing mice via frozen slicing, and the results showed that the folate nanoparticles were preferentially accumulated in the tumor site rather than other tissues, indicating the desired specificity for tumor targeting and imaging. All these findings suggested that this practical synthetic strategy can potentially facilitate the preparation of multifunctional imaging probe in biology and diagnosis of disease.


Assuntos
Diagnóstico por Imagem/métodos , Ácido Fólico/química , Nanopartículas/química , Neoplasias/patologia , Animais , Transplante de Células , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/metabolismo , Células HeLa , Humanos , Teste de Materiais , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias/diagnóstico , Neoplasias/metabolismo , Tamanho da Partícula , Estirenos/química
11.
Zhonghua Zhong Liu Za Zhi ; 29(8): 575-9, 2007 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-18210874

RESUMO

OBJECTIVE: To investigate the possibility of using generation 5 polyamidoamine dendrimers (G5-PAMAM-D) as gene vector for eukaryotic expression plasmid of siRNA in prostate carcinoma in vitro and vivo. METHODS: Firstly, eukaryotic expression vector of siRNA pSilencing 4.1-EGFP-shRNA, specific for enhanced green fluorescent protein (EGFP), pSilencing 4.1-STAT3-shRNA for signal transducers and activators of transcription 3 (STAT3) was constructed. pEGFP-C1 and pSilencing 4.1-EGFP-shRNA were cotransfected into prostate cancer cells PC-3 and 22Rv1 with G5-PAPAM-D as vector, and to observe silencing of EGFP. Next, pSilencing 4.1-STAT3-shRNA was transfected into PC-3 and 22Rv1 cells by G5-PAPAM-D, Western blotting and apoptosis staining was used to detect silencing of STAT3 and growth inhibition. Thirdly, BALB/C mice subcutaneous tumor model was made with PC-3 cells. Polyplex of G5-PAMAM-D and pSilencing 4.1-STAT3-shRNA was injected intratumorally. The tumor volume was measured and recorded. RESULTS: Fluorescence detection and Western blotting analysis demonstrated that G5-PAMAM-D was able to deliver Silencing 4.1-EGFP-shRNA and pSilencing 4.1-STAT3-shRNA into the two prostate cancer cell lines, and shRNA was expressed to induce silence of EGFP and STAT3. MTT results showed that proliferation of prostate cancer cells was suppressed by G5-PAMAM-D/pSilencing 4.1-STAT3-shRNA and induced apoptosis of PC-3 cells in vitro. Human prostate cancer in mice was successfully formed by inoculation of PC-3 cells into male BABL/C mice. In G5-PAMAM-D/pSilencing 4.1-STAT3-shRNA treated group, the tumor volume was shrank remarkably at 9 days after treatment and tumor growth was retarded compared with control groups. CONCLUSION: GS-PAMAM-D nanoparticles can be used to deliver plasmid vector expressing shRNA into prostate cancer cells effectively in vitro and vivo. It appears to be a promising gene vector for RNA interference therapy in prostate cancer.


Assuntos
Proliferação de Células , Inativação Gênica , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Fator de Transcrição STAT3/genética , Animais , Linhagem Celular Tumoral , Dendrímeros , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas , Transplante de Neoplasias , Plasmídeos , Poliaminas/química , Neoplasias da Próstata/metabolismo , Fator de Transcrição STAT3/metabolismo , Transfecção , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...