Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Nanobiotechnology ; 22(1): 178, 2024 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-38614985

RESUMO

BACKGROUND: Clearance of apoptotic cells by efferocytosis is crucial for prevention of atherosclerosis progress, and impaired efferocytosis contributes to the aggravated atherosclerosis. RESULTS: In this study, we found that diabetic ApoE-/- mice showed aggravated atherosclerosis as hyperglycemia damaged the efferocytosis capacity at least partially due to decreased expression of Mer tyrosine kinase (MerTK) on macrophages. To locally restore MerTK in the macrophages in the plaque, hybrid membrane nanovesicles (HMNVs) were thus developed. Briefly, cell membrane from MerTK overexpressing RAW264.7 cell and transferrin receptor (TfR) overexpressing HEK293T cell were mixed with DOPE polymers to produce nanovesicles designated as HMNVs. HMNVs could fuse with the recipient cell membrane and thus increased MerTK in diabetic macrophages, which in turn restored the efferocytosis capacity. Upon intravenous administration into diabetic ApoE-/- mice, superparamagnetic iron oxide nanoparticles (SMN) decorated HMNVs accumulated at the aorta site significantly under magnetic navigation, where the recipient macrophages cleared the apoptotic cells efficiently and thus decreased the inflammation. CONCLUSIONS:  Our study indicates that MerTK decrease in macrophages contributes to the aggravated atherosclerosis in diabetic ApoE-/- mice and regional restoration of MerTK in macrophages of the plaque via HMNVs could be a promising therapeutic approach.


Assuntos
Aterosclerose , Diabetes Mellitus , Humanos , Animais , Camundongos , Eferocitose , Células HEK293 , Membrana Celular , Proteínas Tirosina Quinases , Apolipoproteínas E/genética , Nanopartículas Magnéticas de Óxido de Ferro
2.
Cell Death Differ ; 31(3): 292-308, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38017147

RESUMO

Lipid droplet (LD) accumulation is a notable feature of obesity-induced cardiomyopathy, while underlying mechanism remains poorly understood. Here we show that mice fed with high-fat diet (HFD) exhibited significantly increase in cardiac LD and RTN3 expression, accompanied by cardiac function impairment. Multiple loss- and gain-of function experiments indicate that RTN3 is critical to HFD-induced cardiac LD accumulation. Mechanistically, RTN3 directly bonds with fatty acid binding protein 5 (FABP5) to facilitate the directed transport of fatty acids to endoplasmic reticulum, thereby promoting LD biogenesis in a diacylglycerol acyltransferase 2 dependent way. Moreover, lipid overload-induced RTN3 upregulation is due to increased expression of CCAAT/enhancer binding protein α (C/EBPα), which positively regulates RTN3 transcription by binding to its promoter region. Notably, above findings were verified in the myocardium of obese patients. Our findings suggest that manipulating LD biogenesis by modulating RTN3 may be a potential strategy for treating cardiac dysfunction in obese patients.


Assuntos
Cardiomiopatias , Gotículas Lipídicas , Animais , Camundongos , Proteínas de Transporte/metabolismo , Dieta Hiperlipídica , Proteínas de Ligação a Ácido Graxo/metabolismo , Coração , Gotículas Lipídicas/metabolismo , Lipídeos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Obesidade/metabolismo
3.
Theranostics ; 13(14): 5114-5129, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37771781

RESUMO

Senescent cells in plaques emerge as a detrimental factor for atherosclerosis (AS), for which targeted senolysis might be a promising therapeutic strategy. The development of safe and efficient senolytics for senescent cell eradication by targeted delivery is greatly needed. Methods: Pro-apoptotic intelligent Bax (iBax)-overexpressing plasmid was constructed by molecular cloning, in which Bax CDS was fused to miR-122 recognition sites. Extracellular vesicle-based senolytics (EViTx) were developed to be conjugated with magnetic nanoparticles on the surface, iBax mRNA encapsulated inside, and BAX activator BTSA1 incorporated into the membrane. EViTx was characterized, and in vivo distribution was tracked via fluorescence imaging. The therapeutic effects of EViTx on AS and its systemic side effects were analyzed in ApoE-/- mice. Results: Magnetic nanoparticles, iBax mRNA and BAX activator BTSA1 were efficiently loaded into/onto EViTx. With external magnetic field navigation, EViTx was delivered into atherosclerotic plaques and induced significant apoptosis in senescent cells regardless of origins. Repeated delivery of EViTx via tail vein injection has achieved high therapeutic efficacy in ApoE-/- mice. Notably, EViTx is inevitably accumulated in liver cells, while the iBax mRNA was translationally repressed by miR-122, an endogenous miRNA highly expressed in hepatocytes, and thus the liver cells are protected from the potential toxicity of Bax mRNA. Conclusion: Our work demonstrated that magnetic EV-based delivery of iBax mRNA and the BAX activator BTSA1, efficiently induced apoptosis in recipient senescent cells in atherosclerotic plaques. This strategy represents a promising treatment approach for AS and other age-related diseases.


Assuntos
Aterosclerose , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Vesículas Extracelulares , MicroRNAs , Placa Aterosclerótica , Animais , Camundongos , Placa Aterosclerótica/metabolismo , Proteína X Associada a bcl-2 , Senoterapia , Aterosclerose/terapia , Aterosclerose/metabolismo , Vesículas Extracelulares/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Senescência Celular , RNA Mensageiro/metabolismo
4.
Theranostics ; 13(10): 3467-3479, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37351166

RESUMO

Extracellular vesicle (EV)-based low-density lipoprotein receptor (Ldlr) mRNA delivery showed excellent therapeutic effects in treating familial hypercholesterolemia (FH). Nevertheless, the loading inefficiency of EV-based mRNA delivery presents a significant challenge. Recently, RNA-binding proteins (RBPs) have been fused to EV membrane proteins for selectively encapsulating targeted RNAs to promote loading efficiency. However, the strong interaction between therapeutic RNAs and RBPs prevents RNA release from endosomes to the cytosol in the recipient cells. In this study, an improved strategy was developed for efficient encapsulation of Ldlr mRNA into EVs in donor cells and controllable release in recipient cells. Methods: The MS2 bacteriophage coat protein (CD9-MCP) fusion protein, Ldlr mRNA, and a customized MS2 containing RNA aptamer base-pair matched with Ldlr mRNA were expressed in donor cells. Cells receiving the above therapeutic EVs were simultaneously treated with EVs containing "Ldlr releaser" with a sequence similar to the recognition sites in Ldlr mRNA. Therapeutic effects were analyzed in Ldlr-/- mice receiving EV treatments via the tail vein. Results: In vitro experiments demonstrated improved loading efficiency of Ldlr mRNA in EVs via MS2-MCP interaction. Treatment of "Ldlr releaser" competitively interacted with MS2 aptamer with higher affinity and released Ldlr mRNA from CD9-MCP for efficient translation. When the combinatory EVs were delivered into recipient hepatocytes, the robust LDLR expression afforded therapeutic benefits in Ldlr-/- mice. Conclusion: We proposed an EV-based mRNA delivery strategy for enhanced encapsulation of therapeutic mRNAs in EVs and RNA release into the cytosol for translation in recipient cells with great potential for gene therapy.


Assuntos
Vesículas Extracelulares , Hiperlipoproteinemia Tipo II , Camundongos , Animais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Hiperlipoproteinemia Tipo II/metabolismo , Hepatócitos/metabolismo , RNA/metabolismo , Vesículas Extracelulares/metabolismo
5.
Front Oncol ; 13: 1105482, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37091162

RESUMO

Aims: Abnormal vessel patterns are specific signs in patients with early cervical abnormality and cervical cancer(CC) by colposcopy, but the impact of human papillomavirus (HPV) infections on abnormal vessel patterns remains unknown. Methods: A total of 6716 female patients with HPV infections or cytological abnormalities who underwent a colposcopy following abnormal CC screening results were included in the study. The final pathological diagnosis was confirmed to be the most severe pathological grade across cervical biopsy, endocervical canal curettage (ECC) and conization. Univariate and multivariate logistic regression analyses were used to investigate the association between HPV infections and abnormal vessel patterns, adjusting for age, gravidity and parity. Results: There were 6124 normal vascular cases by colposcopy and 592 cases with cervical vascular abnormality. The prevalence of HPV infections was 4284 (70%) in normal patients, and the prevalence of HPV infections was 479 (80%) in cervical vascular abnormality patients. HPV high-risk type 16 infection alone increased the risk of cervical heteromorphic blood vessels (aOR=3.66, 95%CI: 2.54~5.27). HPV 16 and 33 alone (other than the commonly recognized subtype of 18) or coinfection of these two genotypes could increase the risk of cervical punctate vascular and cervical vascular mosaic features and abnormal cervical blood vessels. An increased risk of abnormal cervical lesions was observed for HPV 16 and 33 alone or combined in coinfection compared to the negative group. The risk of cervical vascular abnormality was increased 10-fold by coinfection with HPV 16 and 33 (aOR=10.67, 95% CI: 4.54~25.09, P<0.001). HPV 16, 33 alone or combined in coinfection were associated with an increased risk of lesions more advanced than high-grade squamous intraepithelial lesion (HSIL) when compared to the negative group. The risk of lesions more advanced than HSIL was up to 26-fold higher in the coinfection with HPV 16 and 33 group than in the negative group (aOR=26.23, 95%CI: 11.23~61.27, P<0.001). Conclusion: HPV16 and 33 are the most dangerous HPV genotypes correlated with abnormal vascular patterns. Combined HPV16 and HPV33 infection increases the risk of abnormal vascular patterns. Combined HPV16 and HPV33 infection increases the risk of developing HSIL+.

6.
Adv Sci (Weinh) ; 10(19): e2301107, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37097746

RESUMO

Cancer vaccine has been considered as a promising immunotherapy by inducing specific anti-tumor immune response. Rational vaccination at suitable time to efficiently present tumor associated antigen will boost tumor immunity and is badly needed. Here, a poly (lactic-co-glycolic acid) (PLGA)-based cancer vaccine of nanoscale is designed, in which engineered tumor cell membrane proteins, mRNAs, and sonosensitizer chlorin e6 (Ce6) are encapsulated at high efficiency. The nanosized vaccine can be efficiently delivered into antigen presentation cells (APCs) in lymph nodes after subcutaneous injection. In the APCs, the encapsulated cell membrane and RNA from engineered cells, which have disturbed splicing resembling the metastatic cells, provide neoantigens of metastatic cancer in advance. Moreover, the sonosensitizer Ce6 together with ultrasound irradiation promotes mRNA escape from endosome, and augments antigen presentation. Through 4T1 syngeneic mouse model, it has been proved that the proposed nanovaccine is efficient to elicit antitumor immunity and thus prevent cancer metastasis.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Camundongos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/metabolismo , Células Dendríticas , RNA/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Apresentação de Antígeno , Membrana Celular/metabolismo , Antígenos de Neoplasias/metabolismo
7.
Adv Sci (Weinh) ; 10(14): e2205692, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36965082

RESUMO

Sepsis is one of the most common causes of death, which is closely related to the uncontrolled systemic inflammation. Dysregulation of M1 macrophage polarization is the primary contributor to serious inflammation. In this study, it is revealed that the murine homologue of circRNA SCAR (steatohepatitis-associated circRNA ATP5B regulator), denoted as circRNA mSCAR hereafter, decreases in the macrophages of septic mice, which correlates with the excessive M1 polarization. To restore circRNA mSCAR in mitochondria, exosomes encapsulated with circRNA mSCAR are further electroporated with poly-D-lysine-graft-triphenylphosphine (TPP-PDL), and thus TPP-PDL facilitates the bound circRNA delivered into mitochondria when the exosomes engulf by the recipient cells. In in vivo septic mouse model and in vitro cell model, it is shown that the exosome-based mitochondria delivery system delivers circRNA mSCAR into mitochondria preferentially in the macrophages, favoring macrophage polarization toward M2 subtype. Accordingly, the systemic inflammation is attenuated by exosome-based mitochondrial delivery of circRNA mSCAR, together with alleviated mortality. Collectively, the results uncover the critical role of circRNA mSCAR in sepsis, and provide a promising approach to attenuate sepsis via exosome-based mitochondrial delivery of circRNA mSCAR.


Assuntos
Exossomos , MicroRNAs , Sepse , Animais , Camundongos , MicroRNAs/genética , RNA Circular/genética , RNA Circular/metabolismo , Exossomos/metabolismo , Ativação de Macrófagos , Inflamação/metabolismo , Sepse/terapia
8.
ACS Appl Mater Interfaces ; 14(45): 50626-50636, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36342824

RESUMO

Exosomes serve as a promising therapeutic nanoplatform. However, the exosomes produced by donor cells are a heterogeneous group, with only a small portion having high therapeutic efficacy. Specific isolation of the subpopulation with high efficacy is important for lowering the dose and minimizing toxicity. In this study, we loaded target mRNA and displayed specific Flag in engineered exosomes simultaneously. Briefly, the donor cells were transfected with plasmid expressing a fusion protein Flag-TCS-PTGFRN-CTSL-MCP, namely, exosome sorter. During biogenesis, the RNA-binding motif MCP can specifically bind with MS2-containing RNA and sort the target RNA into the lumen of exosomes. Anti-Flag magnetic beads can capture and thus purify the engineered exosomes via recognition of the Flag on the surface of exosomes. After purification, the Flag could be cleaved by thrombin treatment while MCP can be separated from the fusion protein by CTSL autocleavage upon exosome acidification, minimizing the side effects and augmenting the therapeutic effects. By the proof-of-concept experiment, the exosome sorter-based "all-in-one" strategy was confirmed effective in both the encapsulation of therapeutic mRNA (Ldlr-MS2) into exosomes and the subsequent purification. The purified Ldlr-MS2-containing exosomes had much higher efficacy in alleviating atherosclerosis, in comparison with the bulk exosomes, confirming the advantage of the proposed "all-in-one" strategy.


Assuntos
Exossomos , Hiperlipoproteinemia Tipo II , Humanos , Exossomos/metabolismo , Hiperlipoproteinemia Tipo II/metabolismo , RNA/metabolismo , RNA Mensageiro/metabolismo
9.
J Nanobiotechnology ; 20(1): 463, 2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36309712

RESUMO

BACKGROUND: Exosome mediated mRNA delivery is a promising strategy for the treatment of multiple diseases. However, the low yield of exosomes is a bottleneck for clinical translation. In this study, we boosted exosome production via simultaneously reducing the expression of genes inhibiting exosome biogenesis and supplementing the culture medium with red cell membrane components. RESULTS: Among the candidate genes, knocking down of Rab4 was identified to have the highest efficacy in promoting exosome biogenesis while without any obvious cytotoxicity. Additionally, supplementing red cell membrane particles (RCMPs) in the culture medium further promoted exosome production. Combination of Rab4 knockdown and RCMP supplement increased exosome yield up to 14-fold. As a proof-of-concept study, low-density lipoprotein receptor (Ldlr) mRNA was forced expressed in the exosome donor cells and passively encapsulated into the exosomes during biogenesis with this strategy. Though exosome production per cell increased, the booster strategy didn't alter the loading efficiency of therapeutic Ldlr mRNA per exosome. Consistently, the therapeutic exosomes derived by the strategy alleviated liver steatosis and atherosclerosis in Ldlr-/- mice, similar as the exosomes produced by routine methods. CONCLUSIONS: Together, the proposed exosome booster strategy conquers the low yield bottleneck to some extent and would certainly facilitate the clinical translation of exosomes.


Assuntos
Exossomos , Camundongos , Animais , Exossomos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Int J Nanomedicine ; 17: 4933-4946, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275481

RESUMO

Background: The early detection of atherosclerotic lesions is particularly important for risk prediction of acute cardiovascular events. Macrophages apoptosis was significantly associated with the degree of AS lesions and especially contributed to plaque vulnerability. In this research, we mainly sought to explore the feasibility of a home-made AV-nanobubbles (NBAV) for visualization of apoptotic macrophages and assessment of atherosclerosis (AS) lesions by contrast-enhanced ultrasound (CEUS) imaging. Methods: NBAV were prepared by "Optimized Thin-Film Hydration" and "Biotin-Avidin-Biotin" methods. Then, the characterization and echogenicity of NBAV were measured and analyzed in vitro. The targeting ability of NBAV to ox-LDL-induced apoptotic macrophages was observed by laser scanning confocal microscope. The ApoE-/- mice mode fed with high fat diet were observed by high-frequency ultrasound, microanatomy and oil red O staining. CEUS imaging in vivo was performed on AS plaques with NBAV and NBCtrl injection through the tail vein in turn in ApoE-/- mice. After CEUS imaging, the plaques were confirmed and analyzed by histopathological and immunological assessment. Results: The prepared NBAV had a nano-scale size distribution with a low PDI and a negative zeta potential. Moreover, NBAV showed an excellent stability and exhibited a significantly echogenic signal than saline in vitro. In addition, we found that NBAV could target apoptotic macrophages induced by ox-LDL. Compared with NBCtrl, CEUS imaging of NBAV showed strong and sustained echo enhancement in plaque area of aortic arch in vivo. Further research showed that NBAV sensitive plaques presented more significant pathological changes with several vulnerable plaque features and abundant TUNEL-positive area. Conclusion: NBAV displayed a sensitive indicator to evaluate apoptotic macrophages, indicating a promising CEUS molecular probe for AS lesions and vulnerable plaques identification.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Camundongos , Avidina , Biotina , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/patologia , Aterosclerose/diagnóstico por imagem , Aterosclerose/patologia , Apolipoproteínas E/genética , Macrófagos/patologia , Sondas Moleculares
11.
Bioact Mater ; 16: 82-94, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35386323

RESUMO

Atherosclerosis is characterized by inflammation in the arterial wall, which is known to be exacerbated by diabetes. Therapeutic repression of inflammation is a promising strategy for treating atherosclerosis. In this study, we showed that diabetes aggravated atherosclerosis in apolipoproteinE knockout (ApoE -/-) mice, in which increased expression of long-chain acyl-CoA synthetase 1 (Acsl1) in macrophages played an important role. Knockdown of Acsl1 in macrophages (Mφ shAcsl1 ) reprogrammed macrophages to an anti-inflammatory phenotype, especially under hyperglycemic conditions. Injection of Mφ shAcsl1 reprogrammed macrophages into streptozotocin (STZ)-induced diabetic ApoE -/- mice (ApoE -/-+ STZ) alleviated inflammation locally in the plaque, liver and spleen. Consistent with the reduction in inflammation, plaques became smaller and more stable after the adoptive transfer of reprogrammed macrophages. Taken together, our findings indicate that increased Acsl1 expression in macrophages play a key role in aggravated atherosclerosis of diabetic mice, possibly by promoting inflammation. Adoptive transfer of Acsl1 silenced macrophages may serve as a potential therapeutic strategy for atherosclerosis.

12.
Bioact Mater ; 8: 494-504, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34541415

RESUMO

Arterial stiffness due to the vessel remodeling is closely linked to raised blood pressure, and its physiopathologic mechanism is still not fully understood. We here aimed to explore whether extracellular vesicle (EV) mediated intercellular communication between endothelium and smooth muscle cell contribute to the blood vessel remodeling under hypertension. We here revealed that the arterial endothelial cells robustly secreted EV, which in turn could be circulated and/or directly taken up by the subendothelial smooth muscle cells (SMC). Under hypertension, the EV secretion increased and the miRNA profile changed significantly mainly due to the raised mechanical force and subsequent enhanced reactive oxygen species generation. Among the miRNA cargos in the EV, miR-320d/423-5p were found increased most significantly. In vivo delivery of miR-320d/423-5p mimics via engineered EV increased their expression in arterial vessels, recapitulating the phenotype in hypertension. In contrast, therapeutic delivery of miR-320d/423-5p inhibitors via engineered EV alleviated the phenotype in spontaneous hypertension rat model. Together, we have found that the injured endothelium due to the raised mechanical force in hypertension contributes to the arterial wall remodeling via the secreted EV. Our study has not only provided novel insights on the mechanism of hypertension associated blood vessel wall remodeling, but also shed light on therapeutic intervention of hypertension associated vascular diseases.

13.
J Nanobiotechnology ; 19(1): 402, 2021 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-34863187

RESUMO

BACKGROUND: Efficient and topical delivery of drugs is essential for maximized efficacy and minimized toxicity. In this study, we aimed to design an exosome-based drug delivery platform endowed with the ability of escaping from phagocytosis at non-target organs and controllably releasing drugs at targeted location. RESULTS: The swtichable stealth coat CP05-TK-mPEG was synthesized and anchored onto exosomes through the interaction between peptide CP05 and exosomal surface marker CD63. Chlorin e6 (Ce6) was loaded into exosomes by direct incubation. Controllable removal of PEG could be achieved by breaking thioketal (TK) through reactive oxygen species (ROS), which was produced by Ce6 under ultrasound irradiation. The whole platform was called SmartExo. The stealth effects were analyzed in RAW264.7 cells and C57BL/6 mice via tracing the exosomes. To confirm the efficacy of the engineered smart exosomes, Bone morphogenetic protein 7 (Bmp7) mRNA was encapsulated into exosomes by transfection of overexpressing plasmid, followed by stealth coating, with the exosomes designated as SmartExo@Bmp7. Therapeutic advantages of SmartExo@Bmp7 were proved by targeted delivering Bmp7 mRNA to omental adipose tissue (OAT) of obese C57BL/6 mice for browning induction. SmartExo platform was successfully constructed without changing the basic characteristics of exosomes. The engineered exosomes effectively escaped from the phagocytosis by RAW264.7 and non-target organs. In addition, the SmartExo could be uptaken locally on-demand by ultrasound mediated removal of the stealth coat. Compared with control exosomes, SmartExo@Bmp7 effectively delivered Bmp7 mRNA into OAT upon ultrasound irradiation, and induced OAT browning, as evidenced by the histology of OAT and increased expression of uncoupling protein 1 (Ucp1). CONCLUSIONS: The proposed SmartExo-based delivery platform, which minimizes side effects and maximizing drug efficacy, offers a novel safe and efficient approach for targeted drug delivery. As a proof, the SmartExo@Bmp7 induced local white adipose tissue browning, and it would be a promising strategy for anti-obesity therapy.


Assuntos
Tecido Adiposo Branco , Proteína Morfogenética Óssea 7 , Sistemas de Liberação de Medicamentos/métodos , RNA Mensageiro , Terapia por Ultrassom , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Administração Tópica , Animais , Bioengenharia , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/farmacocinética , Proteína Morfogenética Óssea 7/farmacologia , Exossomos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/farmacocinética , RNA Mensageiro/farmacologia
14.
Theranostics ; 11(20): 9988-10000, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34815799

RESUMO

Rationale: Tailored inflammation control is badly needed for the treatment of kinds of inflammatory diseases, such as atherosclerosis. IL-10 is a potent anti-inflammatory cytokine, while systemic and repeated delivery could cause detrimental side-effects due to immune repression. In this study, we have developed a nano-system to deliver inflammation-responsive Il-10 mRNA preferentially into macrophages for tailored inflammation control. Methods:Il-10 was engineered to harbor a modified HCV-IRES (hepatitis C virus internal ribosome entry site), in which the two miR-122 recognition sites were replaced by two miR-155 recognition sites. The translational responsiveness of the engineered mRNA to miR-155 was tested by Western blot or ELISA. Moreover, the engineered Il-10 mRNA was passively encapsulated into exosomes by forced expression in donor cells. Therapeutic effects on atherosclerosis and the systemic leaky expression effects in vivo of the functionalized exosomes were analyzed in ApoE-/- (Apolipoprotein E-deficient) mice. Results: The engineered IRES-Il-10 mRNA could be translationally activated in cells when miR-155 was forced expressed or in M1 polarized macrophages with endogenous miR-155 induced. In addition, the engineered IRES-Il-10 mRNA, when encapsulated into the exosomes, could be efficiently delivered into macrophages and some other cell types in the plaque in ApoE-/- mice. In the recipient cells of the plaque, the encapsulated Il-10 mRNA was functionally translated into protein, with relatively low leaky in other tissues/organs without obvious inflammation. Consistent with the robust Il-10 induction in the plaque, exosome-based delivery of the engineered Il-10 could alleviate the atherosclerosis in ApoE-/- mice. Conclusion: Our study established a potent platform for controlled inflammation control via exosome-based systemic and repeated delivery of engineered Il-10 mRNA, which could be a promising strategy for atherosclerosis treatment.


Assuntos
Aterosclerose/terapia , Sistemas de Liberação de Medicamentos/métodos , Interleucina-10/farmacologia , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , China , Exossomos/imunologia , Exossomos/fisiologia , Engenharia Genética/métodos , Células HEK293 , Humanos , Inflamação/metabolismo , Interleucina-10/administração & dosagem , Interleucina-10/genética , Sítios Internos de Entrada Ribossomal/genética , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Camundongos , MicroRNAs/genética , Placa Aterosclerótica/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/farmacologia
15.
Bioact Mater ; 6(9): 2870-2880, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33718668

RESUMO

Immune suppressive microenvironment in tumor emerges as the main obstacle for cancer immunotherapy. In this study, we identified that HIF1α was activated in the tumor associated macrophages and acted as an important factor for the immune suppressive microenvironment. Epigenetically silencing of Hif1α via histone H3 methylation in the promoter region was achieved by CRISPR/dCas9-EZH2 system, in which histone H3 methylase EZH2 was recruited to the promoter region specifically. The Hif1α silenced macrophage, namely HERM (Hif1α Epigenetically Repressed Macrophage) manifested as inheritable tumor suppressing phenotype. In the subcutaneous B16-F10 melanoma syngeneic model, intratumoral injection of HERMs reprogrammed the immune suppressive microenvironment to the active one, reducing tumor burden and prolonging overall survival. Additionally, HERMs therapy remarkably inhibited tumor angiogenesis. Together, our study has not only identified a promising cellular and molecular target for reverting immune suppressive microenvironment, but also provided a potent strategy for reprogramming tumor microenvironment via epigenetically reprogrammed macrophages.

16.
Theranostics ; 11(8): 3996-4010, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664877

RESUMO

Exosomes are nanosized lipid vesicles originating from the endosomal system that carry many macromolecules from their parental cells and play important roles in intercellular communication. The functions and underlying mechanisms of exosomes in atherosclerosis have recently been intensively studied. In this review, we briefly introduce exosome biology and then focus on advances in the roles of exosomes in atherosclerosis, specifically exosomal changes associated with atherosclerosis, their cellular origins and potential functional cargos, and their detailed impacts on recipient cells. We also discuss the potential of exosomes as biomarkers and drug carriers for managing atherosclerosis.


Assuntos
Aterosclerose/etiologia , Exossomos/fisiologia , Aterosclerose/fisiopatologia , Aterosclerose/terapia , Biomarcadores/metabolismo , Plaquetas/fisiologia , Diabetes Mellitus/etiologia , Portadores de Fármacos , Exossomos/genética , Humanos , Metabolismo dos Lipídeos , Macrófagos/fisiologia , MicroRNAs/genética , Modelos Cardiovasculares , Terapia de Alvo Molecular , Obesidade/etiologia , Medicina de Precisão , Fatores de Risco
17.
Placenta ; 105: 85-93, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33556718

RESUMO

INTRODUCTION: There is a strong association between gestational obesity and fetal cardiac dysfunction, while the exact mechanisms remain largely unknown. The purpose of this study was to investigate the role of exosomes from maternal visceral adipose tissue in abnormal embryonic development in obese pregnancy. METHODS: Female C57BL/6J obese mice were induced by a high-fat diet (containing 60% fat). Fetal cardiac function and morphology were examined by echocardiography and histology. The placenta was extracted for histological examination. miRNA expression in exosomes from the visceral adipose tissue was profiled by RNA-seq. Gene expression of inflammatory factors was analyzed by qPCR. RESULTS: In the obese pregnant mice, there were obvious inflammation and lipid droplets in the placenta. And the fetal cardiac function in obese pregnancy was also compromised. Moreover, injection of the visceral adipose tissue exosomes from the obese mice significantly decreased the fetal cardiac function in the normal lean pregnant mice. Mechanistically, the decreased expression of miR-19b might be responsible for the enhanced inflammation in the placenta. DISCUSSION: Exosomes derived from visceral adipose tissue in obese mice contribute to fetal heart dysfunction, at least partially via affecting the function of the placenta.


Assuntos
Exossomos/metabolismo , Coração Fetal/fisiopatologia , Cardiopatias/fisiopatologia , Gordura Intra-Abdominal/metabolismo , Obesidade Materna/fisiopatologia , Animais , Dieta Hiperlipídica , Feminino , Cardiopatias/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade Materna/metabolismo , Placenta/metabolismo , Gravidez
18.
Theranostics ; 11(6): 2953-2965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456582

RESUMO

Familial hypercholesterolemia (FH), with high LDL (low-density lipoprotein) cholesterol levels, is due to inherited mutations in genes, such as low-density lipoprotein receptor (LDLR). Development of therapeutic strategies for FH, which causes atherosclerosis and cardiovascular disease, is urgently needed. Methods: Mice with low-density lipoprotein receptor (Ldlr) deletion (Ldlr-/- mice) were used as an FH model. Ldlr mRNA was encapsulated into exosomes by forced expression of Ldlr in the donor AML12 (alpha mouse liver) cells, and the resultant exosomes were denoted as ExoLdlr. In vivo distribution of exosomes was analyzed by fluorescence labeling and imaging. The delivery efficiency of Ldlr mRNA was analyzed by qPCR and Western blotting. Therapeutic effects of ExoLdlr were examined in Ldlr-/- mice by blood lipids and Oil Red O staining. Results: The encapsulated mRNA was stable and could be translated into functional protein in the recipient cells. Following tail vein injection, exosomes were mainly delivered into the liver, producing abundant LDLR protein, resembling the endogenous expression profile in the wild-type mouse. Compared with control exosomes, ExoLdlr treatment significantly decreased lipid deposition in the liver and lowered the serum LDL-cholesterol level. Significantly, the number and size of atherosclerotic plaques and inflammation were reduced in the ExoLdlr-treated mice. Conclusions: We have shown that exosome-mediated Ldlr mRNA delivery effectively restored receptor expression, treating the disorders in the Ldlr-/- mouse. Our study provided a new therapeutic approach for the treatment of FH patients and managing atherosclerosis.


Assuntos
Exossomos/genética , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Receptores de LDL/genética , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/terapia , Linhagem Celular , Modelos Animais de Doenças , Terapia Genética/métodos , Células HEK293 , Humanos , Hiperlipoproteinemia Tipo II/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/terapia , RNA Mensageiro/genética , Receptores de LDL/metabolismo
19.
J Extracell Vesicles ; 9(1): 1816710, 2020 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-33133429

RESUMO

Therapeutically intervening the function of RNA in vivo remains a big challenge. We here developed an exosome-based strategy to deliver engineered RNA-binding protein for the purpose of recruiting specific RNA to the lysosomes for degradation. As a proof-of-principle study, RNA-binding protein HuR was fused to the C-terminus of Lamp2b, a membrane protein localized in both exosome and lysosome. The fusion protein was able to be incorporated into the exosomes. Moreover, exosomes engineered with Lamp2b-HuR successfully decreased the abundance of RNA targets possibly via lysosome-mediated degradation, especially when the exosomes were acidified. The system was specifically effective in macrophages, which are lysosome enriched and resistant to routine transfection mediated RNAi strategy. In the CCl4-induced liver injury mouse model, we found that delivery of acidified exosomes engineered with Lamp2b-HuR significantly reduced liver fibrosis, together with decreased miR-155 and other inflammatory genes. In summary, the established exosome-based RNA-binding protein delivery strategy, namely "exosome-mediated lysosomal clearance", takes the advantage of exosome in targeted delivery and holds great promise in regulating a set of genes in vivo.

20.
Biochem Biophys Res Commun ; 532(1): 60-67, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-32828538

RESUMO

Chemotherapy related cardiotoxicity is now becoming one of the biggest hurdles for the prognosis of cancer patients. Therapeutically delivering protective small RNAs holds promise for the cardiotoxicity prevention and therapy. However, heart is intrinsically refractory to the nanoparticle-mediated drug delivery. In this study, we found that the exosome-mediated miRNA delivery into the heart could be significantly augmented with the aid of ultrasound targeted microbubble destruction (UTMD). Moreover, we found that UTMD assisted exosomal miR-21 delivery into the heart significantly decreased the cell death, and restored the cardiac function in a doxorubicin induced cardiotoxicity mouse model. Our study here not only provides a promising strategy to protect the heart from the chemotherapy related cardiotoxicity, but also sheds light on gene therapy of other heart diseases.


Assuntos
Cardiotônicos/administração & dosagem , Cardiotoxicidade/prevenção & controle , MicroRNAs/administração & dosagem , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose , Cardiotoxicidade/patologia , Cardiotoxicidade/fisiopatologia , Morte Celular , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Sistemas de Liberação de Medicamentos , Ecocardiografia Doppler de Pulso , Exossomos , Testes de Função Cardíaca , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Microbolhas , Ultrassom
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA