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










Base de dados
Intervalo de ano de publicação
1.
Anticancer Drugs ; 35(5): 397-411, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38527419

RESUMO

This study aimed to investigate the role and molecular mechanism of heme oxygenase-1 (HMOX1) in chemotherapy resistance in small-cell lung cancer (SCLC). Employed bioinformatics, qPCR, and Western Blot to assess HMOX1 levels in SCLC versus normal tissues and its prognostic relevance. CCK-8, flow cytometry, and thiobarbituric acid assays determined HMOX1's impact on SCLC chemosensitivity, ferroptosis markers, lipid peroxidation, and mic14's role in chemoresistance. In the GSE40275 and GSE60052 cohorts, HMOX1 expression was downregulated in SCLC tissues compared to normal tissues. Higher HMOX1 expression was associated with improved prognosis in the Sun Yat-sen University Cancer Hospital cohort and GSE60052 cohort. The RNA and protein levels of HMOX1 were reduced in drug-resistant SCLC cell lines compared to chemosensitive cell lines. Upregulation of HMOX1 increased chemosensitivity and reduced drug resistance in SCLC, while downregulation of HMOX1 decreased chemosensitivity and increased drug resistance. Upregulation of HMOX1 elevated the expression of ferroptosis-related proteins ACSL4, CD71, Transferrin, Ferritin Heavy Chain, and Ferritin Light Chain, while decreasing the expression of GPX4 and xCT. Conversely, downregulation of HMOX1 decreased the expression of ACSL4, CD71, Transferrin, Ferritin Heavy Chain, and Ferritin Light Chain, while increasing the expression of GPX4 and xCT. Upregulation of HMOX1 promoted cellular lipid peroxidation, whereas downregulation of HMOX1 inhibited cellular lipid peroxidation. Upregulation of HMOX1 reduced the RNA level of mic14, while downregulation of HMOX1 increased the RNA level of mic14. mic14 exhibited inhibitory effects on cellular lipid peroxidation in SCLC cells and contributed to reduced chemosensitivity and increased drug resistance in chemoresistant SCLC cell lines. HMOX1 plays a role in ferroptosis by regulating mic14 expression, thereby reversing chemoresistance in SCLC.


Assuntos
Ferroptose , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Apoferritinas/genética , Apoferritinas/farmacologia , Apoferritinas/uso terapêutico , Heme Oxigenase-1/genética , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo , RNA/farmacologia , RNA/uso terapêutico , Transferrinas/farmacologia
2.
Angew Chem Int Ed Engl ; 62(22): e202302255, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-36959091

RESUMO

Ferrous iron (Fe2+ ) has more potent hydroxyl radical (⋅OH)-generating ability than other Fenton-type metal ions, making Fe-based nanomaterials attractive for chemodynamic therapy (CDT). However, because Fe2+ can be converted by ferritin heavy chain (FHC) to nontoxic ferric form and then sequestered in ferritin, therapeutic outcomes of Fe-mediated CDT agents are still far from satisfactory. Here we report the synthesis of siRNA-embedded Fe0 nanoparticles (Fe0 -siRNA NPs) for self-reinforcing CDT via FHC downregulation. Upon internalization by cancer cells, pH-responsive Fe0 -siRNA NPs are degraded to release Fe2+ and FHC siRNA in acidic endo/lysosomes with the aid of oxygen (O2 ). The accompanied O2 depletion causes an intracellular pH decrease, which further promotes the degradation of Fe0 -siRNA NPs. In addition to initiating chemodynamic process, Fe2+ -catalyzed ⋅OH generation facilitates endo/lysosomal escape of siRNA by disrupting the membranes, enabling FHC downregulation-enhanced CDT.


Assuntos
Nanopartículas , Neoplasias , Humanos , Ferro/metabolismo , Apoferritinas/metabolismo , Apoferritinas/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Regulação para Baixo , Radical Hidroxila/metabolismo , Nanopartículas/uso terapêutico , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Peróxido de Hidrogênio/metabolismo
3.
Bioconjug Chem ; 34(5): 845-855, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-36827653

RESUMO

Biological nanoparticles, such as proteins and extracellular vesicles, are rapidly growing as nanobased drug-delivery agents due to their biocompatibility, high loading efficiency, and bioavailability. However, most of the candidates emerging preclinically hardly confirm their potential when entering clinical trials. Among other reasons, this is due to the low control of synthesis processes and the limited characterization of their potential immunoreactivity profiles. Here, we propose a combined method that allow us to fully characterize H-ferritin nanoparticles' immunoreactivity during their production, purification, endotoxin removal, and drug loading. H-Ferritin is an extremely interesting nanocage that is being under evaluation for cancer therapy due to its innate cancer tropism, favorable size, and high stability. However, being a recombinant protein, its immunoreactivity should be carefully evaluated preclinically to enable further clinical translation. Surprisingly, this aspect is often underestimated by the scientific community. By measuring proinflammatory cytokine release as a function of endotoxin content, we found that even removing all pyrogenic contaminants from the nanocage, a mild immunoreactivity was still left. When we further purified H-ferritin by loading doxorubicin through a highly standardized loading method, proinflammatory cytokine release was eliminated. This confirmed the safety of H-ferritin nanocages to be used for drug delivery in cancer therapy. Our approach demonstrated that when evaluating the safety of nanodrugs, a combined analysis of acute toxicity and immunoreactivity is necessary to guarantee the safety of newly developed products and to unveil their real translational potential.


Assuntos
Nanopartículas , Neoplasias , Humanos , Apoferritinas/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Citocinas/uso terapêutico
4.
Acta Biomater ; 160: 265-280, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36822483

RESUMO

Myocardial ischemia-reperfusion injury (MI/RI) seriously restricts the therapeutic effect of reperfusion. It is demonstrated that ferroptosis and apoptosis of cardiomyocytes are widely involved in MI/RI. Therefore, simultaneous inhibition of ferroptosis and apoptosis of cardiomyocytes can be a promising strategy to treat MI/RI. Besides, transferrin receptor 1 (TfR1) is highly expressed in ischemic myocardium, and apoferritin (ApoFn) is a ligand of the transferrin receptor. In this study, CsA@ApoFn was prepared by wrapping cyclosporin A (CsA) with ApoFn and actively accumulated in ischemic cardiomyocytes through TfR1 mediated endoctosis in MI/RI mice. After entering cardiomyocytes, ApoFn in CsA@ApoFn inhibited ferroptosis of ischemic cardiomyocytes by increasing the protein expression of GPX4 and reducing the content of labile iron pool and lipid peroxides. At the same time, CsA in CsA@ApoFn attenuated the apoptosis of ischemic cardiomyocytes through recovering mitochondrial membrane potential and reducing the level of reactive oxygen species, which played a synergistic role with ApoFn in the treatment of MI/RI. In conclusion, CsA@ApoFn restored cardiac function of MI/RI mice by simultaneously blocking ferroptosis and apoptosis of cardiomyocytes. ApoFn itself not only served as a safe carrier to specifically deliver CsA to ischemic cardiomyocytes but also played a therapeutic role on MI/RI. CsA@ApoFn is proved as an effective drug delivery platform for the treatment of MI/RI. STATEMENT OF SIGNIFICANCE: Recent studies have shown that ferroptosis is an important mechanism of myocardial ischemia-reperfusion injury (MI/RI). Therefore, simultaneous inhibition of ferroptosis and apoptosis of cardiomyocytes can be a promising strategy to treat MI/RI. Apoferritin, as a delivery carrier, can actively target to ischemic myocardium through binding with highly expressed transferrin receptor on ischemic cardiomyocytes. At the same time, apoferritin plays a protective role on ischemic cardiomyocytes by inhibiting ferroptosis. This strategy of killing two birds with one stone significantly improves the therapeutic effect on MI/RI while does not need more pharmaceutical excipients, which has the prospect of clinical transformation.


Assuntos
Ferroptose , Traumatismo por Reperfusão Miocárdica , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/metabolismo , Ciclosporina/farmacologia , Ciclosporina/química , Ciclosporina/metabolismo , Apoferritinas/farmacologia , Apoferritinas/metabolismo , Apoferritinas/uso terapêutico , Apoptose
5.
Lab Invest ; 102(7): 741-752, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35351965

RESUMO

Invasive growth of glioblastoma makes residual tumor unremovable by surgery and leads to disease relapse. Temozolomide is widely used first-line chemotherapy drug to treat glioma patients, but development of temozolomide resistance is almost inevitable. Ferroptosis, an iron-dependent form of non-apoptotic cell death, is found to be related to temozolomide response of gliomas. However, whether inducing ferroptosis could affect invasive growth of glioblastoma cells and which ferroptosis-related regulators were involved in temozolomide resistance are still unclear. In this study, we treated glioblastoma cells with RSL3, a ferroptosis inducer, in vitro (cell lines) and in vivo (subcutaneous and orthotopic animal models). The treated glioblastoma cells with wild-type or mutant IDH1 were subjected to RNA sequencing for transcriptomic profiling. We then analyze data from our RNA sequencing and public TCGA glioma database to identify ferroptosis-related biomarkers for prediction of prognosis and temozolomide resistance in gliomas. Analysis of transcriptome data from RSL3-treated glioblastoma cells suggested that RSL3 could inhibit glioblastoma cell growth and suppress expression of genes involved in cell cycle. RSL3 effectively reduced mobility of glioblastoma cells through downregulation of critical genes involved in epithelial-mesenchymal transition. Moreover, RSL3 in combination with temozolomide showed suppressive efficacy on glioblastoma cell growth, providing a promising therapeutic strategy for glioblastoma treatment. Although temozolomide attenuated invasion of glioblastoma cells with mutant IDH1 more than those with wild-type IDH1, the combination of RSL3 and temozolomide similarly impaired invasive ability of glioblastoma cells in spite of IDH1 status. Finally, we noticed that both ferritin heavy chain 1 and ferritin light chain predicted unfavorable prognosis of glioma patients and were significantly correlated with mRNA levels of methylguanine methyltransferase as well as temozolomide resistance. Altogether, our study provided rationale for combination of RSL3 with temozolomide to suppress glioblastoma cells and revealed ferritin heavy chain 1 and ferritin light chain as biomarkers to predict prognosis and temozolomide resistance of glioma patients.


Assuntos
Neoplasias Encefálicas , Ferroptose , Glioblastoma , Glioma , Animais , Apoferritinas/farmacologia , Apoferritinas/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Temozolomida/farmacologia , Temozolomida/uso terapêutico
6.
Mol Cancer Ther ; 21(4): 535-545, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35131878

RESUMO

High frequency of KRAS and TP53 mutations is a unique genetic feature of pancreatic ductal adenocarcinoma (PDAC). TP53 mutation not only renders PDAC resistance to chemotherapies but also drives PDAC invasiveness. Therapies targeting activating mutant KRAS are not available and the outcomes of current PDAC treatment are extremely poor. Here, we report that MMRi62, initially identified as an MDM2-MDM4-targeting small molecule with p53-independent pro-apoptotic activity, shows anti-PDAC activity in vitro and in vivo. We show that MMRi62 inhibits proliferation, clonogenic, and spheroid growth of PDAC cells by induction of cell death. MMRi62-induced cell death in PDAC is characteristic of ferroptosis that is associated with increased autophagy, increased reactive oxygen species, and lysosomal degradation of NCOA4 and ferritin heavy chain (FTH1). In addition to induced degradation of FTH1, MMRi62 also induces proteasomal degradation of mutant p53. Interestingly, MMRi62-induced ferroptosis occurs in PDAC cell lines harboring either KRAS and TP53 double mutations or single TP53 mutation. In orthotopic xenograft PDAC mouse models, MMRi62 was capable of inhibiting tumor growth in mice associated with downregulation of NCOA4 and mutant p53 in vivo. Strikingly, MMRi62 completely abrogated metastasis of orthotopic tumors to distant organs, which is consistent with MMRi62's ability to inhibit cell migration and invasion in vitro. These findings identified MMRi62 as a novel ferroptosis inducer capable of suppressing PDAC growth and overcoming metastasis.


Assuntos
Carcinoma Ductal Pancreático , Ferroptose , Neoplasias Pancreáticas , Animais , Apoferritinas/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/genética
7.
Theranostics ; 12(4): 1800-1815, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35198074

RESUMO

Rationale: With the advantages of tumor-targeting, pH-responsive drug releasing, and biocompatibility, ferritin nanocage emerges as a promising drug carrier. However, its wide applications were significantly hindered by the low loading efficiency of hydrophobic drugs. Herein, we redesigned the inner surface of ferritin drug carrier (ins-FDC) by fusing the C- terminus of human H ferritin (HFn) subunit with optimized hydrophobic peptides. Methods: Hydrophobic and hydrophilic drugs were encapsulated into the ins-FDC through the urea-dependent disassembly/reassembly strategy and the natural drug entry channel of the protein nanocage. The morphology and drug loading/releasing abilities of the drug-loaded nanocarrier were then examined. Its tumor targeting character, system toxicity, application in synergistic therapy, and anti-tumor action were further investigated. Results: After optimization, 39 hydrophobic Camptothecin and 150 hydrophilic Epirubicin were encapsulated onto one ins-FDC nanocage. The ins-FDC nanocage exhibited programed drug release pattern and increased the stability and biocompatibility of the loaded drugs. Furthermore, the ins-FDC possesses tumor targeting property due to the intrinsic CD71-binding ability of HFn. The loaded drugs may penetrate the brain blood barrier and accumulate in tumors in vivo more efficiently. As a result, the drugs loaded on ins-FDC showed reduced side effects and significantly enhanced efficacy against glioma, metastatic liver cancer, and chemo-resistant breast tumors. Conclusions: The ins-FDC nanocarrier offers a promising novel means for the delivery of hydrophobic compounds in cancer treatments, especially for the combination therapies that use both hydrophobic and hydrophilic chemotherapeutics.


Assuntos
Ferritinas , Glioma , Apoferritinas/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Epirubicina , Ferritinas/química , Glioma/tratamento farmacológico , Humanos
8.
Nanoscale ; 13(3): 1875-1883, 2021 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-33439183

RESUMO

Conventionally, a disassembly and reassembly method has been used for encapsulation of drug molecules in ferritin protein nano-cages. However, clinical applications of ferritin have been greatly restricted by its limited drug-loading capacity and process complexity. Here, we establish a simple high yield process for preparing high drug-loaded ferritin nanomedicine for industrial production. A complex of ferritin and a target drug was obtained by incubating the mixture at an appropriate pH. An electrostatic charge potential and small ferritin cavity facilitates the passage of drug molecules through the pores, traversing the ferritin shell and enabling deposition of the drug in the ferritin cavity. Compared to the disassembly/reassembly method, the loading capacity of a doxorubicin-loaded ferritin heavy chain (DOX-FTH), constructed by our novel method, was over 3-fold higher, while doxorubicin recovery was 10-fold higher. Results of transmission electron microscopy, size exclusion chromatography, dynamic light scattering, and zeta potential indicate that DOX-FTH exhibits the same physicochemical characteristics of natural apo-ferritin. Moreover, DOX-FTH can be taken up and induce apoptosis of cancer cells overexpressing TfR1. Here, we have demonstrated the successful introduction of more than ten drug molecule types into ferritin nano-cages using a novel method. These results demonstrate that this one-step method is a powerful production process to construct a drug-loading ferritin drug delivery system carrier.


Assuntos
Neoplasias , Preparações Farmacêuticas , Apoferritinas/uso terapêutico , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Ferritinas , Neoplasias/tratamento farmacológico
9.
J Hematol Oncol ; 13(1): 123, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32928251

RESUMO

BACKGROUND: Conventional therapeutic approaches for tumor angiogenesis, which are primarily focused on the inhibition of active angiogenesis to starve cancerous cells, target the vascular endothelial growth factor signaling pathway. This aggravates hypoxia within the tumor core and ultimately leads to increased tumor proliferation and metastasis. To overcome this limitation, we developed nanoparticles with antiseptic activity that target tumor vascular abnormalities. METHODS: Ferritin-based protein C nanoparticles (PCNs), known as TFG and TFMG, were generated and tested in Lewis lung carcinoma (LLC) allograft and MMTV-PyMT spontaneous breast cancer models. Immunohistochemical analysis was performed on tumor samples to evaluate the tumor vasculature. Western blot and permeability assays were used to explore the role and mechanism of the antitumor effects of PCNs in vivo. For knocking down proteins of interest, endothelial cells were transfected with siRNAs. Statistical analysis was performed using one-way ANOVA followed by post hoc Dunnett's multiple comparison test. RESULTS: PCNs significantly inhibited hypoxia and increased pericyte coverage, leading to the inhibition of tumor growth and metastasis, while increasing survival in LLC allograft and MMTV-PyMT spontaneous breast cancer models. The coadministration of cisplatin with PCNs induced a synergistic suppression of tumor growth by improving drug delivery as evidenced by increased blood prefusion and decreased vascular permeability. Moreover, PCNs altered the immune cell profiles within the tumor by increasing cytotoxic T cells and M1-like macrophages with antitumor activity. PCNs induced PAR-1/PAR-3 heterodimerization through EPCR occupation and PAR-1 activation, which resulted in Gα13-RhoA-mediated-Tie2 activation and stabilized vascular tight junctions via the Akt-FoxO3a signaling pathway. CONCLUSIONS: Cancer treatment targeting the tumor vasculature by inducing antitumor immune responses and enhancing the delivery of a chemotherapeutic agent with PCNs resulted in tumor regression and may provide an effective therapeutic strategy.


Assuntos
Apoferritinas/uso terapêutico , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Nanopartículas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Proteína C/uso terapêutico , Receptor TIE-2/fisiologia , Remodelação Vascular/efeitos dos fármacos , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/uso terapêutico , Apoferritinas/administração & dosagem , Bevacizumab/uso terapêutico , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Hipóxia Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Cisplatino/uso terapêutico , Técnicas de Cocultura , Sistemas de Liberação de Medicamentos , Sinergismo Farmacológico , Células Endoteliais/efeitos dos fármacos , Feminino , Masculino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Neoplasias/fisiologia , Pericitos/metabolismo , Proteína C/administração & dosagem , Organismos Livres de Patógenos Específicos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
10.
Nanoscale ; 11(26): 12449-12459, 2019 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-31231742

RESUMO

Ferritin, a natural iron storage protein, is endowed with a unique structure, the ability to self-assemble and excellent physicochemical properties. Beyond these, genetic manipulation can easily tune the structure and functions of ferritin nanocages, which further expands the biomedical applications of ferritin. Here, we focus on human H-ferritin, a recently discovered ligand of transferrin receptor 1, to review its derived variants and related structures and properties. We hope this review will provide new insights into how to rationally design versatile protein cage nanocarriers for effective disease treatment.


Assuntos
Apoferritinas , Portadores de Fármacos , Ferro/química , Nanopartículas , Apoferritinas/química , Apoferritinas/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico
11.
Theranostics ; 9(8): 2167-2182, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31149036

RESUMO

Hepatocellular carcinoma (HCC) remains one of the leading causes of cancer deaths, primarily due to its high incidence of recurrence and metastasis. Considerable efforts have therefore been undertaken to develop effective therapies; however, effective anti-HCC therapies rely on identification of suitable biomarkers, few of which are currently available for drug targeting. Methods: GRP78 was identified as the membrane receptor of HCC-targeted peptide SP94 by immunoprecipitation and mass spectrum analysis. To develop an effective anti-HCC drug nanocarrier, we first displayed GRP78-targeted peptide SP94 onto the exterior surface of Pyrococcus furiosus ferritin Fn (HccFn) by genetic engineering approach, and then loaded doxorubicin (Dox) into the cavities of HccFn via urea-based disassembly/reassembly method, thereby constructing a drug nanocarrier called HccFn-Dox. Results: We demonstrated that HccFn nanocage encapsulated ultra-high dose of Dox (up to 400 molecules Dox/protein nanocage). In vivo animal experiments showed that Dox encapsulated in HccFn-Dox was selectively delivered into HCC tumor cells, and effectively killed subcutaneous and lung metastatic HCC tumors. In addition, HccFn-Dox significantly reduced drug exposure to healthy organs and improved the maximum tolerated dose by six-fold compared with free Dox. Conclusion: In conclusion, our findings clearly demonstrate that GRP78 is an effective biomarker for HCC therapy, and GRP78-targeted HccFn nanocage is effective in delivering anti-HCC drug without damage to healthy tissue.


Assuntos
Antineoplásicos/administração & dosagem , Apoferritinas/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Doxorrubicina/análogos & derivados , Proteínas de Choque Térmico/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Nanopartículas/química , Células 3T3 , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Apoferritinas/farmacocinética , Apoferritinas/uso terapêutico , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos , Chaperona BiP do Retículo Endoplasmático , Feminino , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ligação Proteica , Distribuição Tecidual
12.
Int J Mol Sci ; 20(10)2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31100837

RESUMO

Ferritin is a spherical iron storage protein composed of 24 subunits and an iron core. Using biomimetic mineralization, magnetic iron oxide can be synthesized in the cavity of ferritin to form magnetoferritin (MFt). MFt, also known as a superparamagnetic protein, is a novel magnetic nanomaterial with good biocompatibility and flexibility for biomedical applications. Recently, it has been demonstrated that MFt had tumor targetability and a peroxidase-like catalytic activity. Thus, MFt, with its many unique properties, provides a powerful platform for tumor diagnosis and therapy. In this review, we discuss the biomimetic synthesis and biomedical applications of MFt.


Assuntos
Apoferritinas/metabolismo , Apoferritinas/uso terapêutico , Ferro/metabolismo , Ferro/uso terapêutico , Óxidos/metabolismo , Óxidos/uso terapêutico , Animais , Fenômenos Biomecânicos , Meios de Contraste , Sistemas de Liberação de Medicamentos , Compostos Férricos , Ferritinas , Humanos , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/patologia
13.
Sci Rep ; 8(1): 8867, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29891921

RESUMO

Herein, we describe the in vivo effects of doxorubicin (DOX) encapsulated in ubiquitous protein apoferritin (APO) and its efficiency and safety in anti-tumor treatment. APODOX is both passively (through Enhanced Permeability and Retention effect) and actively targeted to tumors through prostate-specific membrane antigen (PSMA) via mouse antibodies conjugated to the surface of horse spleen APO. To achieve site-directed conjugation of the antibodies, a HWRGWVC heptapeptide linker was used. The prostate cancer-targeted and non-targeted nanocarriers were tested using subcutaneously implanted LNCaP cells in athymic mice models, and compared to free DOX. Prostate cancer-targeted APODOX retained the high potency of DOX in attenuation of tumors (with 55% decrease in tumor volume after 3 weeks of treatment). DOX and non-targeted APODOX treatment caused damage to liver, kidney and heart tissues. In contrast, no elevation in liver or kidney enzymes and negligible changes were revealed by histological assessment in prostate cancer-targeted APODOX-treated mice. Overall, we show that the APO nanocarrier provides an easy encapsulation protocol, reliable targeting, high therapeutic efficiency and very low off-target toxicity, and is thus a promising delivery system for translation into clinical use.


Assuntos
Apoferritinas/uso terapêutico , Doxorrubicina/análogos & derivados , Imunoconjugados/uso terapêutico , Nanoconjugados/uso terapêutico , Neoplasias da Próstata/terapia , Animais , Antígenos de Superfície/imunologia , Apoferritinas/efeitos adversos , Linhagem Celular Tumoral , Doxorrubicina/efeitos adversos , Doxorrubicina/uso terapêutico , Glutamato Carboxipeptidase II/imunologia , Coração/efeitos dos fármacos , Xenoenxertos , Humanos , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias da Próstata/tratamento farmacológico , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Bioconjug Chem ; 29(4): 1384-1398, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29505243

RESUMO

Recently, nanoparticles (NPs) have been widely investigated for delivery of anticancer drugs. Here, a dual control drug-release modality was developed that uses naturally occurring protein apoferritin loaded with doxorubicin (DOX) and ADS-780 near-infrared (NIR) fluorescent dye-decorated NPs (ADNIR NPs). ADNIR NPs act as a grenade to detonate the targeted tumor site following laser irradiation (photothermal therapy, PTT) and explode into cluster warheads (apoferritin-loaded DOX nanocages, AF-DOX NCs) that further destroy the tumor cells (chemotherapy). Light was shown to disrupt the grenade-like structure of NPs to release AF-DOX NCs as well as DOX from NCs in low-pH intercellular environments. In vitro and in vivo studies showed that the structure of AF-DOX NCs was disassembled to release DOX, which then killed the cancer cells in organelles with acidic environments. In vivo studies showed that the ADNIR NP-decorated with NIR dye facilitated tracking of the accumulated NPs at the tumor site using an IVIS imaging system. Overall, targeted ADNIR NPs with dual-release mechanisms were developed for use in photothermal theranostic and chemotherapy. This modality has high potential for application in cancer treatment and clinical translation for drug delivery and imaging.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/terapia , Doxorrubicina/uso terapêutico , Corantes Fluorescentes/uso terapêutico , Nanopartículas/uso terapêutico , Nanomedicina Teranóstica/métodos , Animais , Antibióticos Antineoplásicos/administração & dosagem , Apoferritinas/administração & dosagem , Apoferritinas/uso terapêutico , Neoplasias do Colo/patologia , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/uso terapêutico , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Feminino , Corantes Fluorescentes/administração & dosagem , Células HT29 , Humanos , Hipertermia Induzida/métodos , Raios Infravermelhos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Imagem Óptica/métodos , Fototerapia/métodos
15.
Nanomedicine (Lond) ; 9(14): 2233-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25405799

RESUMO

Nanomedicine as a continuously evolving discipline is still looking for a structure with perfect properties that is usable as a multifunctional transporter. Great potential is attributed to synthetic materials such as fullerenes, porous hollow silica nanoparticles and single-wall nanotubes, among others. However, materials that are natural to the human body are more acceptable by the organism, and thus become an attractive approach in this field of research. Ferritins are proteins that naturally occur in most living organisms throughout evolution and may be a possible transporter choice. Numerous applications have demonstrated the possibilities of iron-free ferritins, called apoferritins, serving as platforms for various nanomedical purposes This article summarizes the advantages and disadvantages of these proteins and discusses their practical applications and future perspectives.


Assuntos
Apoferritinas/uso terapêutico , Nanomedicina , Apoferritinas/química , Técnicas Biossensoriais , Portadores de Fármacos , Terapia Genética , Humanos , Conformação Proteica
16.
Proc Natl Acad Sci U S A ; 111(41): 14900-5, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25267615

RESUMO

An ideal nanocarrier for efficient drug delivery must be able to target specific cells and carry high doses of therapeutic drugs and should also exhibit optimized physicochemical properties and biocompatibility. However, it is a tremendous challenge to engineer all of the above characteristics into a single carrier particle. Here, we show that natural H-ferritin (HFn) nanocages can carry high doses of doxorubicin (Dox) for tumor-specific targeting and killing without any targeting ligand functionalization or property modulation. Dox-loaded HFn (HFn-Dox) specifically bound and subsequently internalized into tumor cells via interaction with overexpressed transferrin receptor 1 and released Dox in the lysosomes. In vivo in the mouse, HFn-Dox exhibited more than 10-fold higher intratumoral drug concentration than free Dox and significantly inhibited tumor growth after a single-dose injection. Importantly, HFn-Dox displayed an excellent safety profile that significantly reduced healthy organ drug exposure and improved the maximum tolerated dose by fourfold compared with free Dox. Moreover, because the HFn nanocarrier has well-defined morphology and does not need any ligand modification or property modulation it can be easily produced with high purity and yield, which are requirements for drugs used in clinical trials. Thus, these unique properties make the HFn nanocage an ideal vehicle for efficient anticancer drug delivery.


Assuntos
Apoferritinas/uso terapêutico , Doxorrubicina/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoferritinas/farmacocinética , Apoferritinas/farmacologia , Relação Dose-Resposta a Droga , Doxorrubicina/sangue , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Endocitose/efeitos dos fármacos , Feminino , Células HT29 , Humanos , Injeções , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/ultraestrutura , Neoplasias/sangue , Neoplasias/patologia , Distribuição Tecidual/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Artigo em Inglês | MEDLINE | ID: mdl-23606622

RESUMO

Ferritin, a major iron storage protein found in most living organisms, is composed of a 24-subunit protein cage with a hollow interior cavity. Serum ferritin serves as a critical marker to detect total body iron status. However, recent research reveals a number of novel functions of ferritin besides iron storage; for example, a ferritin receptor, transferrin receptor 1 (TfR1), has been identified and serum ferritin levels are found to be elevated in tumors. A particular new finding is that magnetoferritin nanoparticles, biomimetically synthesized using H-chain ferritin to form a 24-subunit cage with an iron oxide core, possess intrinsic dual functionality, the protein shell specifically targeting tumors and the iron oxide core catalyzing peroxidase substrates to produce a color reaction allowing visualization of tumor tissues. Here we attempt to summarize current research on ferritin, particularly newly identified functions related to tumors, in order to address current challenges and highlight future directions.


Assuntos
Ferritinas/uso terapêutico , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Apoferritinas/uso terapêutico , Biomarcadores Tumorais/metabolismo , Sistemas de Liberação de Medicamentos , Ferritinas/química , Ferritinas/metabolismo , Humanos , Ferro/uso terapêutico , Proteínas de Ligação ao Ferro/metabolismo , Óxidos/uso terapêutico , Receptores de Superfície Celular/metabolismo
18.
Small ; 8(16): 2505-14, 2012 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-22619186

RESUMO

Human ferritin H-chain protein (FTH1)-based nanoparticles possess a precisely assembled nanometer-scale structure and high safety. However, their applications for imaging and drug delivery towards cancer cells remain limited due to a lack of target specificity. Epidermal growth factor receptor (EGFR) is overexpressed in many malignant tissues including breast cancer, and has been used as a therapeutic target for cancer treatment. Herein, a genetic method is shown to generate EGF-FTH1 chimeric proteins. EGF-FTH1 nanoparticles with EGF on the surface are then produced. The data demonstrate that EGF-FTH1 nanoparticles, with a small size (11.8 ± 1.8 nm), narrow size distribution, and high biosafety, can specifically bind to and then be taken up by breast cancer MCF-7 cells and MDA-MB-231 cells, but not normal breast epithelial MCF-10A cells. In contrast, binding and absorption of nontargeted ferritin-based nanoparticles to breast cancer cells are negligible. In vivo studies show that EGF-FTH1 nanoparticles are accumulated in breast tumors in a mouse xenograft model. Interestingly, the concentration of EGF-FTH1 nanoparticles in the tumor site is significantly reduced when mice are pretreated with an excess of free EGF. These results imply that EGF-EGFR interaction plays an important role in regulating the tumor retention of EGF-FTH1 nanoparticles.


Assuntos
Apoferritinas/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Fator de Crescimento Epidérmico/uso terapêutico , Nanopartículas/uso terapêutico , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Luz , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/efeitos adversos , Nanopartículas/ultraestrutura , Ligação Proteica , Proteínas Recombinantes de Fusão/isolamento & purificação , Espalhamento de Radiação , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Proc Natl Acad Sci U S A ; 89(22): 11064-8, 1992 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-1438316

RESUMO

A method is described to deliver 235U to tumors; the isotope would then be fissioned by incident neutrons, producing localized lethal radiation sufficient for therapy. Apoferritin was loaded with an average of approximately 800 238U atoms per molecule. Stability of the loaded apoferritin in solution was improved, so that only 8% loss of uranium occurred after 8 days at pH 7. Fab' antibody fragments were covalently attached to the uranium-loaded apoferritin, and the immunoreactivity of the conjugate was 92% of that for antibody alone. Such bio-uranium constructions should provide significant advantages over boronated antibodies to meet the requirements for clinical neutron-capture therapy.


Assuntos
Apoferritinas/uso terapêutico , Imunoglobulina G/uso terapêutico , Neoplasias/radioterapia , Terapia por Captura de Nêutron/métodos , Urânio/uso terapêutico , Terapia por Captura de Nêutron de Boro/métodos , Estabilidade de Medicamentos , Humanos , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Cinética , Terapia por Captura de Nêutron/instrumentação
20.
J Lab Clin Med ; 96(3): 470-9, 1980 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6995540

RESUMO

We studied the effect of PGE1 in pharmacologic doses on immune complex-induced glomerulonephritis produced by daily intraperitoneal injections of apoferritin. Mice received one of the following injection schedules: apoferritin 4 mg/day, apoferritin 4 mg/day plus PGE1 200 microgram twice daily, saline, or PGE1 200 microgram twice daily. Administration of apoferritin alone resulted in mesangial cell proliferation in all 14 mice with crescent formation in nine. Evidence of subepithelial and mesangial immune complex deposition and a significant increase in urine protein excretion was found. Treatment with PGE1 resulted in a mild increase in mesangial cells in six of 14 mice. No mice developed crescents on this regimen. In addition, proteinuria was prevented, and there was a marked diminution of immune complex deposition. Antiapoferritin antibody was detected in the sera of mice from both groups. No alteration in lymphocyte response to mitogen or in vitro PGE1 suppression of blastogenesis was detected. Our results indicate that PGE1 therapy alters immune complex glomerulonephritis in this model of murine chronic serum sickness by reducing glomerular immune complex deposition. However, no difference in specific or nonspecific immunologic responses was detected.


Assuntos
Prostaglandinas E/uso terapêutico , Doença do Soro/tratamento farmacológico , Animais , Anticorpos , Apoferritinas/imunologia , Apoferritinas/uso terapêutico , Divisão Celular , Doença Crônica , Imunofluorescência , Imunoglobulina G , Imunoglobulina M , Glomérulos Renais/patologia , Glomérulos Renais/ultraestrutura , Linfócitos/citologia , Masculino , Camundongos , Proteinúria/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...