Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Heliyon ; 9(6): e16870, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37346322

RESUMO

Background: Prostate cancer (PCa) is among the most generally diagnosed cancers in males. A long non-coding RNA (lncRNA) called AC245100.4 has been discovered and linked to PCa carcinogenesis. However, its specific and potential mechanism is uncertain in PCa. In this research, we investigated the role of AC245100.4 in cell proliferation and the underlying mechanism in PCa cells. Methods: qRT-PCR assays were utilized to detect AC245100.4 expression and confirm its downstream target. The pathways related to AC245100.4 were identified by RAP-MS. PCa cell proliferation was experimented by Cell Counting Kit-8 and Colony formation assays. Western blot was performed to detect PAR2, AKT, p-AKT, Cyclin D1 and PCNA expression. Results: AC245100.4/PAR2 overexpression promotes PCa cell proliferation and the opposite results are obtained after AC245100.4/PAR2 knockdown. Mechanistically, we found that PAR2 is confirmed as the AC245100.4 downstream target and AC245100.4 promotes PCa cell proliferation by regulating PAR2. AC245100.4 promotes PCa cell proliferation via PI3K/AKT pathway. Rescue assays validated that PAR2 knockdown reversed the impact of AC245100.4 overexpression on increasing p-AKT protein levels. Conclusion: This research revealed that AC245100.4 enhances cell proliferation in PCa cells through modulating the PAR2/PI3K/AKT axis, which may offer novel tumor markers and potential therapeutic targets for PCa.

2.
Bioact Mater ; 25: 569-579, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37056257

RESUMO

Crosstalk between Kupffer cells (KCs) and hepatic stellate cells (HSCs) plays an important role in multiple liver disease conditions, including the formation of liver fibrosis in alcohol-associated liver disease (AALD). Therapeutic targeting of the KC-HSC crosstalk is a prime target for therapeutic interventions. Herein, a novel modular nanosystem was designed and prepared through the self-assembly utilizing boric acid and catechol interactions to prepare polymers modified with a CXCR4-inhibiting moieties. The polymers were used to encapsulate anti-miR-155 and to block the undesirable crosstalk between HSCs and KCs by downregulating miR-155 expression in KCs with the parallel inhibition of CXCR4 signaling in activated HSCs. The combined inhibition of miR-155 and CXCR4 at two different liver cell types achieved improved antifibrosis effects in a mouse model of AALD fibrosis. Our finding highlights the key role that blocking the undesirable crosstalk between HSCs and KCs plays in reversing AALD fibrosis as well as demonstrates a proof-of-concept approach for designing and constructing multifunctional delivery nanosystems using orthogonal functional modules based on the understanding of disease mechanisms.

3.
Biomater Adv ; 136: 212755, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35929290

RESUMO

RNA interference (RNAi) is an emerging therapeutic modality for cancer, which remains in critical need of effective delivery vectors due to the unfavorable biopharmaceutical properties of small RNAs. Polyamines are essential for functioning of mammalian cells. Dysregulated polyamine metabolism is found in many cancers and has been an attractive therapeutic target in combination therapies. Combination therapies based on drugs that affect polyamine metabolism and nucleic acids promise to enhance anticancer activity due to a cooperative effect on multiple oncogenic pathways. Here, we report bioactive polycationic prodrug (F-PaP) based on an anticancer polyamine analogue bisethylnorspermine (BENSpm) modified with perfluoroalkyl moieties. Following encapsulation of siRNA, F-PaP/siRNA nanoparticles were coated with hyaluronic acid (HA) to form ternary nanoparticles HA@F-PaP/siRNA. The presence of perfluoroalkyl moieties and HA reduced cell membrane toxicity and improved stability of the particles with cooperatively enhanced siRNA delivery in pancreatic and colon cancer cell lines. We then tested a therapeutic hypothesis that combining BENSpm with siRNA silencing of polo-like kinase 1 (PLK1) would result in cooperative cancer cell killing. HA@F-PaP/siPLK1 induced polyamine catabolism and cell cycle arrest, leading to enhanced apoptosis in the tested cell lines. The HA-coated nanoparticles facilitated tumor accumulation and contributed to strong tumor inhibition and favorable modulation of the immune tumor microenvironment in orthotopic pancreatic cancer model.


Assuntos
Fluorocarbonos , Neoplasias Pancreáticas , Pró-Fármacos , Animais , Ácido Hialurônico , Mamíferos/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Poliaminas/metabolismo , Pró-Fármacos/farmacologia , RNA Interferente Pequeno/metabolismo , Microambiente Tumoral
4.
Biomater Adv ; 1362022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35813988

RESUMO

RNA interference (RNAi) is an emerging therapeutic modality for cancer, which remains in critical need of effective delivery vectors due to the unfavorable biopharmaceutical properties of small RNAs. Polyamines are essential for functioning of mammalian cells. Dysregulated polyamine metabolism is found in many cancers and has been an attractive therapeutic target in combination therapies. Combination therapies based on drugs that affect polyamine metabolism and nucleic acids promise to enhance anticancer activity due to a cooperative effect on multiple oncogenic pathways. Here, we report bioactive polycationic prodrug (F-PaP) based on an anticancer polyamine analog bisethylnorspermine (BENSpm) modified with perfluoroalkyl moieties. Following encapsulation of siRNA, F-PaP/siRNA nanoparticles were coated with hyaluronic acid (HA) to form ternary nanoparticles HA@F-PaP/siRNA. The presence of perfluoroalkyl moieties and HA reduced cell membrane toxicity and improved stability of the particles with cooperatively enhanced siRNA delivery in pancreatic and colon cancer cell lines. We then tested a therapeutic hypothesis that combining BENSpm with siRNA silencing of polo-like kinase 1 (PLK1) would result in cooperative cancer cell killing. HA@F-PaP/siPLK1 induced polyamine catabolism and cell cycle arrest, leading to enhanced apoptosis in the tested cell lines. The HA-coated nanoparticles facilitated tumor accumulation and contributed to strong tumor inhibition and favorable modulation of the immune tumor microenvironment in orthotopic pancreatic cancer model. Combination anticancer therapy with polyamine prodrug-mediated delivery of siRNA. Hyaluronate coating of the siRNA nanoparticles facilitates selective accumulation in orthotopic pancreatic tumors. Perfluoroalkyl conjugation reduces toxicity and improves gene silencing effect. Nanoparticle treatment induces polyamine catabolism and cell cycle arrest leading to strong tumor inhibition and favorable modulation of immune tumor microenvironment.

5.
Carbohydr Polym ; 294: 119777, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35868792

RESUMO

Aspidopterys obcordata vine is a Chinese Dai ethnic herb used to treat urolithiasis. However, the material basis and underlying mechanisms remain undefined. In this study, a 2.3 kD inulin-like A. obcordata fructan (AOFOS) was isolated by size exclusion column chromatography and characterized by ultrahigh-performance liquid chromatography-ion trap-time of flight mass spectrometry (UPLC-IT-TOF-MS), nuclear magnetic resonance (NMR) spectroscopy, gas chromatography mass spectrometry (GC-MS) and high-performance gel permeation chromatography (HGPC). In addition, AOFOS showed unique anti-urolithiasis activity in Drosophila kidney stone models. Mechanism study indicated that AOFOS reduced the size of calcium oxalate crystals by inhibiting the formation of large size crystals and the generation rate of calcium oxalate crystals as well as the crystal form conversion from calcium oxalate monohydrate (COM) to calcium oxalate dihydrate (COD).


Assuntos
Cálculos Renais , Malpighiaceae , Oxalato de Cálcio/química , Cristalização , Frutanos , Inulina , Cálculos Renais/química
6.
Med Oncol ; 39(5): 94, 2022 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-35570225

RESUMO

Prostate cancer (PCa) is the second most common cause of cancer-related mortality in men. Prostate cancer metastasis usually observed at the last stage is the major cause of prostate cancer-related death. Long non-coding RNAs were reported to be involved in tumorigenesis and progression of prostate cancer. This study aimed to investigate the effects and related mechanisms of AC245100.4 in prostate cancer. Knockdown and overexpression of AC245100.4 was used to detect the effect of AC245100.4 on cell migration. qRT-PCR was used to confirm the downstream target of AC245100.4. RAP-MS was used to find pathways related to AC245100.4. Western blot was performed to detect the expression of p-p38 and p38. We found that AC245100.4 promoted the migration of prostate cancer cells via regulating PAR2. The AC245100.4 or PAR2 knockdown resulted in a decrease in Vimentin but an increase in E-cadherin protein levels, while the AC245100.4 or PAR2 overexpression got the opposite results. Moreover, we discovered that AC245100.4 activated the p38-MAPK via regulating PAR2. In brief, these results have suggested that AC245100.4 and PAR2 served as oncogenic factors in cellular migration in PCa cells.


Assuntos
Neoplasias da Próstata , RNA Longo não Codificante , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/patologia , RNA Longo não Codificante/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia
7.
Cell Biol Int ; 46(7): 1156-1168, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35293661

RESUMO

The Apaf-1 interacting protein (APIP), a ubiquitously expressed antiapoptotic molecule, is aberrantly expressed and of great significance in various cancers. However, little is known regarding the potential value and underlying mechanisms of APIP in prostate cancer. Here, we demonstrated that APIP expression is significantly upregulated in prostate cancer cell lines. APIP overexpression promoted tumor cell proliferation and migration and induced extracellular regulated protein kinases 1/2 (ERK1/2) activation. Pharmacological inhibition of ERK1/2 signaling reversed APIP-induced increase in cell proliferation and migration induced by APIP overexpression. Expression of APIP was hampered by miR-146a-3p. A dual luciferase reporter gene assay identified the regulatory relationship between APIP and miR-146a-3p in prostate cancer, suggesting that APIP is a direct target of miR-146a-3p. miR-146a-3p reduced cell proliferation and migration in prostate cancer. Furthermore, miR-146a-3p inhibited ERK1/2 activation. Application of an ERK1/2 inhibitor reversed the increase in cell proliferation and migration induced by miR-146a-3p inhibition. In summary, this study focused on the role of APIP in regulating cell growth and migration and proposes a theoretical basis for APIP as a promising biomarker in prostate cancer development.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/metabolismo , MicroRNAs , Neoplasias da Próstata , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Quinases/metabolismo
8.
ACS Appl Mater Interfaces ; 14(8): 10015-10029, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35188730

RESUMO

Pancreatic cancer (PC) is a fatal human cancer, whose progression is highly dependent on the nervous tumor microenvironment. In the present study, cationic perfluorocarbon nanoemulsions were employed as an intraperitoneal delivery platform to facilitate the delivery and penetration of a therapeutic small interfering RNA (siRNA) to orthotopic pancreatic tumors. The nanoemulsion was used to silence the expression of the nerve growth factor (NGF) as a way of favorably modulating the tumor-neuronal interactions in pancreatic tumors. The nanoemulsions exhibited deep tumor penetration that was dependent on exocytosis and enhanced NGF gene silencing in vitro and in vivo when compared with control polycation/siRNA polyplexes, leading to the effective and safe suppression of tumor growth in orthotopic PC. Overall, emulsion-assisted delivery of NGF siRNA is a promising treatment approach for PC by targeting the interactions between the tumor cells and the nervous microenvironment.


Assuntos
Neoplasias Pancreáticas , Microambiente Tumoral , Linhagem Celular Tumoral , Inativação Gênica , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , RNA Interferente Pequeno/genética
9.
Epigenomics ; 13(20): 1591-1605, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34657447

RESUMO

Aim: To explore the role and mechanism of long noncoding RNA AC245100.4 and NR4A3 in prostate cancer (PCa). Methods: RNA-sequencing analysis was used to detect the downstream genes of AC245100.4. A series of gain- and loss-of-function approaches were used to investigate the roles of AC245100.4 and NR4A3. RNA immunoprecipitation was performed to examine the interaction between AC245100.4 and STAT3. Results: AC245100.4 was significantly upregulated in PCa cells and tissues. Knockdown of AC21500.4 significantly inhibited the tumorigenesis of PCa cells. Mechanistically, AC245100.4 deregulated the transcription of NR4A3 via increasing p-STAT3, which acted as a transcriptional repressor of NR4A3. Conclusion: Knockdown of long noncoding RNA AC245100.4 inhibits the tumorigenesis of PCa cells via the STAT3/NR4A3 axis.


Lay abstract Long noncoding RNA has recently gained attention for the vital role it plays in the mechanism of prostate cancer (PCa). In this study, the authors found that long noncoding RNA AC245100.4 inhibited the tumor formation of PCa cells via the STAT3/NR4A3 axis. A deeper understanding of the specific mechanism of AC245100.4 in PCa tumor formation will provide insights into diagnostic and prognostic strategies for PCa.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/genética , Neoplasias da Próstata/etiologia , RNA Longo não Codificante/genética , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/genética , Fator de Transcrição STAT3/genética , Animais , Apoptose , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Masculino , Camundongos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
10.
Biochem Biophys Res Commun ; 562: 119-126, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34051575

RESUMO

BACKGROUND: Prostate cancer (PCa) refers to malignant tumors derived from prostate epithelial cells, whose morbidity and mortality rates have been increasing every year. Although new drugs for treating prostate cancer continue to emerge, the unclear mechanism underlying drug targets limits this therapy, thereby constraining identification of effective therapeutic targets. Although GDP dissociation inhibitor 2(GDI2) is highly expressed and closely associated with occurrence and development of many tumors, its role in prostate cancer remains unclear. In this study, we investigated the role of GDI2 and elucidated its underlying mechanism of action in prostate cancer. Moreover, we screened chemotherapeutic drugs that affect GDI2 expression with a view of identifying novel targets for diagnosis and treatment of prostate cancer. METHODS: We performed sequence analyses and functional assays to precisely elucidate the GDI2 role in prostate cancer. Moreover, we induced tumorigenesis in nude mice to verify the role of GDI2 in vivo. Finally, we used the CCK8 assay to ascertain the most suitable IC50 across the three drugs and performed quantitative real time polymerase chain reaction (qRT-PCR) and Western Blot to analyze the effects of drugs on expression of GDI2, p75NTR, and p-NFκB. RESULTS: GDI2 was up-regulated in prostate cancer cells and tissues. Knocking down GDI2 suppressed cell proliferation but promoted cell apoptosis. Interestingly, knocking down GDI2 activated the p75NTR signaling pathway, indicating, for the first time, that p75NTR is negatively correlated with GDI2 expression. CONCLUSION: Taken together, these results indicate that GDI2 is a therapeutic target of paclitaxel. Knocking down of GDI2 inhibits cell proliferation and promotes cell apoptosis via the p75NTR signaling pathway in prostate cancer. Notably, paclitaxel inhibits GDI2 expression, implying that GDI2 may be a promising therapeutic target in prostate cancer.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Paclitaxel/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/genética , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
11.
Sensors (Basel) ; 19(24)2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31835900

RESUMO

The opacity of conventional ultrasound transducers can impede the miniaturization and workflow of current photoacoustic systems. In particular, optical-resolution photoacoustic microscopy (OR-PAM) requires the coaxial alignment of optical illumination and acoustic-detection paths through complex beam combiners and a thick coupling medium. To overcome these hurdles, we developed a novel OR-PAM method on the basis of our recently reported transparent lithium niobate (LiNbO3) ultrasound transducer (Dangi et al., Optics Letters, 2019), which was centered at 13 MHz ultrasound frequency with 60% photoacoustic bandwidth. To test the feasibility of wearable OR-PAM, optical-only raster scanning of focused light through a transducer was performed while the transducer was fixed above the imaging subject. Imaging experiments on resolution targets and carbon fibers demonstrated a lateral resolution of 8.5 µm. Further, we demonstrated vasculature mapping using chicken embryos and melanoma depth profiling using tissue phantoms. In conclusion, the proposed OR-PAM system using a low-cost transparent LiNbO3 window transducer has a promising future in wearable and high-throughput imaging applications, e.g., integration with conventional optical microscopy to enable a multimodal microscopy platform capable of ultrasound stimulation.

12.
ACS Appl Mater Interfaces ; 11(20): 18123-18132, 2019 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-31026135

RESUMO

In situ injectable hydrogels hold great potential for in vivo applications such as drug delivery and regenerative medicine. However, it is challenging to ensure stable sequestration and sustained release of loaded biomolecules in these hydrogels. As aptamers have high binding affinities and specificities against target biomolecules, we studied the capability of aptamers in functionalizing in situ injectable fibrin (Fn) hydrogels for in vivo delivery of two growth factors including vascular endothelial growth factor (VEGF) and platelet-derived growth factor-BB (PDGF-BB). The results show that aptamer-functionalized fibrinogen (Fg) could form in situ injectable Fn hydrogels with porous structures. The aptamer-functionalized Fn hydrogels could sequester more VEGF and PDGF-BB than the native Fn and release these growth factors in a sustained manner with high bioactivity. After the aptamer-functionalized Fn hydrogels were subcutaneously injected into mice, the codelivery of VEGF and PDGF-BB could promote the growth of mature blood vessels. Therefore, this study has successfully demonstrated that aptamer-functionalized in situ injectable hydrogels hold great potential for in vivo codelivery of multiple growth factors and promotion of angiogenesis .


Assuntos
Aptâmeros de Nucleotídeos , Becaplermina , Fibrina , Hidrogéis , Neovascularização Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular , Animais , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Becaplermina/química , Becaplermina/farmacologia , Feminino , Fibrina/química , Fibrina/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/farmacologia
13.
ACS Appl Mater Interfaces ; 11(3): 2880-2890, 2019 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-30592403

RESUMO

Drug-loaded injectable hydrogels have been proven to possess huge potential for applications in tissue engineering. However, increasing the drug loading capacity and regulating the release system to adapt to the microenvironment after myocardial infarction face a huge challenge. In this research, an ROS-sensitive injectable hydrogel strengthened by self-nanodrugs was constructed. A hyperbranched ROS-sensitive macromer (HB-PBAE) with multiacrylate end groups was synthesized through dynamic controlled Michael addition. Meanwhile, a simple protocol based on dopamine polymerization was employed to generate a polydopamine (PDA) layer deposited on the tanshinone IIA (TIIA) nanoparticles (NPs) formed from spontaneous hydrophobic self-assembly. The HB-PBAE reacted with thiolate-modified hyaluronic acid (HA-SH) to form an in situ hydrogel, where TIIA@PDA NPs can be conveniently entrapped through the chemical cross-link between thiolate and quinone groups on PDA, which doubles the modulus of hydrogels. The in vivo degradation behavior of the hydrogels was characterized by MRI, exhibiting a much slower degradation behavior that is markedly different from that of in vitro. Importantly, a significant improvement of cardiac functions was achieved after hydrogel injection in terms of increased ejection fraction and decreased infarction size, accompanied by inhibition of the expression of inflammation factors, such as IL-1ß, IL-6, and TNF-α.


Assuntos
Hidrogéis/administração & dosagem , Inflamação/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Nanopartículas/administração & dosagem , Engenharia Tecidual , Abietanos/administração & dosagem , Abietanos/química , Animais , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Ácido Hialurônico/química , Hidrogéis/química , Indóis/administração & dosagem , Indóis/química , Inflamação/diagnóstico por imagem , Inflamação/genética , Inflamação/fisiopatologia , Interleucina-1beta/genética , Interleucina-6/genética , Imageamento por Ressonância Magnética , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Nanopartículas/química , Polímeros/administração & dosagem , Polímeros/química , Coelhos , Espécies Reativas de Oxigênio/química , Fator de Necrose Tumoral alfa/genética
14.
Biomater Sci ; 6(12): 3300-3308, 2018 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-30350829

RESUMO

Nature-inspired nanoparticles, from pathogens to mammalian cells, have attracted increasing attention, for their specific functions and unparalled features that are often desired in designing drug/gene delivery nonviral vectors. However, the applications of nonviral vectors are still suffering from the limits of low drug loading efficiency and/or low gene transfection efficiency. Herein, a novel carrier-free nanodrug-based virus-surface-mimicking gene delivery nanosystem is designed by condensing doxorubicin nanoparticles (DNPs) onto the surface of the PEI/DNA nanocomplex through electrostatic force, which would prolong the blood circulation time of PEI/DNA and confer high drug loading characteristics to the PEI/DNA nanosystem. Meanwhile, the gene transfection efficiency of DNA can also be enhanced for the increased roughness of coated DNPs. The in vitro and in vivo results demonstrate that carrier-free nanodrug-based gene delivery nanosystems with high drug loading efficiency (97.5%) as well as a rough surface can enhance the endocytosis of the nanoparticles, and consequently enhance the chemo/gene co-therapy of cancers. This is the first time soft materials are used to design virus-surface-mimicking nanocarriers, avoiding the side effects of inorganic materials caused by their non-degradable property. Importantly, our delicate design opens a new pathway to develop nature-inspired nanoparticles for cancer synergistic therapy.


Assuntos
Portadores de Fármacos/química , Endocitose , Técnicas de Transferência de Genes , Nanopartículas/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Células COS , Chlorocebus aethiops , DNA/química , DNA/genética , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Portadores de Fármacos/farmacocinética , Células HeLa , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/metabolismo
15.
Adv Sci (Weinh) ; 5(7): 1800155, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30027047

RESUMO

Carrier-free nanoparticles with high drug loading have attracted increasing attention; however, in situ on-demand drug release remains a challenge. Here, a novel near-infrared (NIR) laser-induced blasting carrier-free nanodrug delivery system is designed and fabricated by coating doxorubicin (DOX) nanoparticles (DNPs) with a polydopamine film (PDA) that would prolong the blood circulation time of DNPs and avoid the preleakage of the DOX during blood circulation. Meanwhile, the NH4HCO3 is introduced to trigger in situ "bomb-like" release of DOX for the production of carbon dioxide (CO2) and ammonia (NH3) gases driven by NIR irradiated photothermal effect of PDA. Both in vitro and in vivo studies demonstrate that the carrier-free nanovectors with high drug loading efficiency (85.8%) prolong tumor accumulation, enhance chemotherapy, achieve the synergistic treatment of chemotherapy and photothermal treatment, and do not induce any foreign-body reaction over a three-week implantation. Hence, the delicate design opens a self-assembly path to develop PDA-based NIR-responsive multifunctional carrier-free nanoparticles for tumor therapy.

16.
Cell Mol Biol (Noisy-le-grand) ; 64(3): 103-107, 2018 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-29506638

RESUMO

Atopic dermatitis (AD) is one of the most prevalent skin diseases around the world. Excessive histamine plays a critical role as an inflammatory factor in the pathogenesis of AD. Deregulated microRNAs (miRNAs) were involved in atopic dermatitis by targeting various genes. MiR-223 had been reported to play a vital role in hematopoiesis. In this study, we identified upregulated miR-223 in the whole blood cells of a large group of AD patients. What's more, we found for the first time that one of the major histamine degradation enzymes, histamine-N-methyltransferase (HNMT), was increased in AD patients and AD model mice. Although there was one miR-223 binding site in the 3'- untranslated region of the HNMT gene, HNMT were not inhibited by miR-223. Taken together, it suggested that miR-223 participates in AD through upregulating HNMT indirectly to degrade the excessive histamine.


Assuntos
Dermatite Atópica/genética , Histamina N-Metiltransferase/genética , MicroRNAs/genética , Regulação para Cima , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Feminino , Células HEK293 , Células Hep G2 , Histamina/metabolismo , Histamina N-Metiltransferase/metabolismo , Humanos , Lactente , Masculino , Camundongos Endogâmicos C57BL , Adulto Jovem
17.
Biomaterials ; 160: 69-81, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29396380

RESUMO

Myocardial infarction (MI) leads to the mass death of cardiomyocytes accompanying with the unfavorable alternation of microenvironment, a fibrosis scar deprived of electrical communications, and the lack of blood supply in the infarcted myocardium. The three factors are inextricably intertwined and thus result in a conservative MI therapy efficacy in clinic. A holistic approach pertinently targeted to these three key points would be favorable to rebuild the heart functions. Here, an injectable conductive hydrogel was constructed via in situ Michael addition reaction between multi-armed conductive crosslinker tetraaniline-polyethylene glycol diacrylate (TA-PEG) and thiolated hyaluronic acid (HA-SH). The resultant soft conductive hydrogel with equivalent myocardial conductivity and anti-fatigue performance was loaded with plasmid DNA encoding eNOs (endothelial nitric oxide synthase) nanocomplexes and adipose derived stem cells (ADSCs) for treating MI. The TA-PEG/HA-SH/ADSCs/Gene hydrogel-based holistic system was injected into the infarcted myocardium of SD rats. We demonstrated an increased expression of eNOs in myocardial tissue the heightening of nitrite concentration, accompanied with upregulation of proangiogenic growth factors and myocardium related mRNA. The results of electrocardiography, cardiogram, and histological analysis convincingly revealed a distinct increase of ejection fraction (EF), shortened QRS interval, smaller infarction size, less fibrosis area, and higher vessel density, indicating a significant improvement of heart functions. This conception of combination approach by a conductive injectable hydrogel loaded with stem cells and gene-encoding eNOs nanoparticles will become a robust therapeutic strategy for the treatment of MI.


Assuntos
Portadores de Fármacos , Hidrogéis , Infarto do Miocárdio/tratamento farmacológico , Óxido Nítrico Sintase Tipo III , Plasmídeos , Animais , Terapia Genética/métodos , Humanos , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/uso terapêutico , Plasmídeos/genética , Plasmídeos/uso terapêutico , Ratos , Ratos Sprague-Dawley
18.
Biomaterials ; 159: 25-36, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29309991

RESUMO

Cell membrane-camouflaged nanoparticles for cancer therapy have received a burgeoning interest over the past years. However, the low drug loading and intratumoral release efficiency, and lack of precise targeting remains a big challenge; in addition, foreign carriers used may pose an expected burden in the course of metabolism. In this study, we designed and fabricated a novel NIR-responsive highly targeted carrier-free nanosystem by coating the exactly identical source of cracked cancer cell membranes (CCCMs) specifically derived from the homologous tumors onto the surface of the co-assembly nanoparticles of doxorubicin (DOX) and FDA-approved photothermal agent, indocyanine green (ICG). The nanosystems exhibited a high drug loading capacity (89.8%), cancer cell self-recognized ability and immune escape function. Further, the nanodrugs could be efficiently released for the membrane disturbance triggered by photothermal effect of ICG under NIR irradiation. The tumor-bearing mice model demonstrated that the self-carried DOX NPs@ICG@CCCM nanoparticles possessed a strong synergistic chemo-/photothermal therapeutic efficacy against tumors in vivo. The present strategy could be developed as a universal approach for designing and constructing carrier-free theranostic nanovehicles by intentionally selecting specific cancer cell membrane and the inner loading cargoes.


Assuntos
Doxorrubicina/química , Nanopartículas/química , Fototerapia/métodos , Animais , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Difusão Dinâmica da Luz , Feminino , Citometria de Fluxo , Células HeLa , Humanos , Hipertermia Induzida/métodos , Verde de Indocianina/química , Camundongos , Microscopia Confocal , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/terapia
19.
Mol Med Rep ; 16(3): 3687-3693, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28765931

RESUMO

Acute myelocytic leukemia (AML) is the most common type of acute leukemia. Long non­coding RNAs (lncRNAs) serve an important role in regulating gene expression through chromatin modification, transcription and post­transcriptional processing. LncRNA H19 was considered as an independent prognostic marker for patients with tumors. The expression of lncRNA H19 was identified to be significantly upregulated in bone marrow samples from patients with AML­M2. Furthermore, it was demonstrated that the knockdown of lncRNA H19 resulted in increased expression of hsa­microRNA (miR)­19a/b and decreased expression of inhibitor of DNA binding 2 (ID2) in AML cells. The knockdown of lncRNA H19 inhibited the proliferation of AML cells in vitro, which could be partially reversed by ID2 overexpression. Furthermore, the results of the bioinformatic analysis revealed potential hsa­miR­19a/b­3p binding sites in lncRNA H19 and ID2. Altogether, the results of the present study suggest that lncRNA H19 regulates the expression of ID2 through competitive binding to hsa­miR­19a and hsa­miR­19b, which may serve a role in AML cell proliferation.


Assuntos
Regulação Leucêmica da Expressão Gênica , Proteína 2 Inibidora de Diferenciação/biossíntese , Leucemia Mieloide Aguda/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Células HL-60 , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética
20.
Front Mol Neurosci ; 10: 27, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28223918

RESUMO

Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease. Recent studies employing microRNA-seq and genome-wide sequencing have identified some non-coding RNAs that are influentially involved in AD pathogenesis. Non-coding RNAs can compete with other endogenous RNAs by microRNA response elements (MREs) and manipulate biological processes, such as tumorigenesis. However, only a few non-coding RNAs have been reported in the pathogenesis of AD. In this study, we constructed the first competing endogenous RNA (ceRNA) network leveraging whole transcriptome sequencing and a previously studied microRNA-seq of APPswe/PS1ΔE9 transgenic mice. The underlying mechanisms for the involvement of ceRNA in AD were validated using the Dual Luciferase Reporter Assay, detection of transcription levels by quantitative RT-PCR and translation levels by Western blotting, and morphological examination in primary cultured neurons. In the ceRNA network, four lncRNAs (C030034L19Rik, Rpph1, A830012C17Rik, and Gm15477) and five miRNAs (miR-182-5p, miR-330-5p, miR-326-3p, miR-132-3p, and miR-484) are enriched in nine pathways and an AD-related gene pool. Among them, Ribonuclease P RNA component H1 (Rpph1) is upregulated in the cortex of APPswe/PS1ΔE9 mice compared to wild type controls. Rpph1 binds to miR326-3p/miR-330-5p and causes the release of their downstream target Cdc42, which leads to CDC42 upregulation. This effect was disrupted upon mutation of the MRE on Rpph1. Moreover, overexpression of Rpph1 increased dendritic spine density in primary cultured hippocampal pyramidal neurons, whereas knocking down of Rpph1 had the reverse effect. In conclusion, Rpph1 modulates CDC42 expression level in a ceRNA-dependent manner, which may represent a compensatory mechanism in the early stage of the AD pathogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA