Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Curr Gene Ther ; 20(5): 321-332, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32998676

RESUMO

Adeno-associated virus (AAV) is a promising vector for in vivo gene therapy because of its excellent safety profile and ability to mediate stable gene expression in human subjects. However, there are still numerous challenges that need to be resolved before this gene delivery vehicle is used in clinical applications, such as the inability of AAV to effectively target specific tissues, preexisting neutralizing antibodies in human populations, and a limited AAV packaging capacity. Over the past two decades, much genetic modification work has been performed with the AAV capsid gene, resulting in a large number of variants with modified characteristics, rendering AAV a versatile vector for more efficient gene therapy applications for different genetic diseases.


Assuntos
Proteínas do Capsídeo/genética , Dependovirus/genética , Doenças Genéticas Inatas/terapia , Terapia Genética/tendências , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/uso terapêutico , Dependovirus/imunologia , Doenças Genéticas Inatas/genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos
2.
Arch Iran Med ; 23(9): 593-599, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32979905

RESUMO

BACKGROUND: Selective therapy has always been the main challenge in cancer treatments. Various non-replicative oncolytic viral systems have revealed the safety and efficacy of using viruses and these products. The aim of this paper is to examine the impact of recombinant apoptin on the proliferation of lung cancer and breast cancer cell lines. METHODS: The present study consisted of two steps of expression of recombinant apoptin and its anti-proliferative effects on normal and cancer cells. In the first step, following bioinformatics and optimizing apoptin gene sequencing and synthesis, it was expressed using vector PET28a and E. coli BL21 (DE3). The expressed recombinant apoptin was confirmed by analytical SDSPAGE and then purified using Ni affinity chromatography. In the second step, the antiproliferative effects of recombinant apoptin on lung cancer, breast cancer and primary cell lines were determined using MTT assay. RESULTS: According to the results of SDS-PAGE gel assay, recombinant apoptin was visible in the 14 kDa band. Also, the MTT assay results indicated that the antiproliferative effects of recombinant apoptin in cancer cell lines was different compared with the primary cell line, and followed a dose-dependent manner in both cell lines. The highest cytotoxicity (lowest cell viability) groups were 0.2 mg/mL in lung cancer (0.32 ± 0.015) (P<0.001), and in breast cancer (0.33 ± 0.031) (P<0.001) and 0.032 mg/mL in primary cells (0.17 ± 0.004) (P<0.01), as compared to the control groups. CONCLUSION: Our results confirmed that recombinant apoptin can induce antiproliferative effects in lung cancer and breast cancer cell lines, but not in normal monkey kidney cell line Vero; thus, it can be introduced as a promising novel specific antitumor agent after further evaluation in clinical trials.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proteínas do Capsídeo/uso terapêutico , Vírus da Anemia da Galinha/genética , Neoplasias Pulmonares/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/isolamento & purificação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Vírus da Anemia da Galinha/metabolismo , Chlorocebus aethiops , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Terapia Viral Oncolítica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/uso terapêutico , Células Vero
3.
Theranostics ; 10(7): 2930-2942, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194845

RESUMO

Goals of investigation: The 5-year survival rate for pancreatic ductal adenocarcinoma (PDAC) has remained at <5% for decades because no effective therapies have been identified. Integrin αvß6 is overexpressed in most PDAC and represents a promising therapeutic target. Thus, we attempted to develop an αvß6-specific peptide-drug conjugate (PDC) for therapy of PDAC. Methodology: We conjugated the DNA-binding pyrrolobenzodiazepine (PBD)-based payload SG3249 (tesirine) to an αvß6-specific 20mer peptide from the VP1 coat protein of foot-and-mouth-disease virus (FMDV) (forming conjugate SG3299) or to a non-targeting peptide (forming conjugate SG3511). PDCs were tested for specificity and toxicity on αvß6-negative versus-positive PDAC cells, patient-derived cell lines from tumor xenografts, and on two different in vivo models of PDAC. Immunohistochemical analyses were performed to establish therapeutic mechanism. Results: The αvß6-targeted PDC SG3299 was significantly more toxic (up to 78-fold) for αvß6-expressing versus αvß6-negative PDAC cell lines in vitro, and achieved significantly higher toxicity at equal dose than the non-targeted PDC SG3511 (up to 15-fold better). Moreover, SG3299 eliminated established (100mm3) Capan-1 PDAC human xenografts, extending the lifespan of mice significantly (P=0.005). Immunohistochemistry revealed SG3299 induced DNA damage and apoptosis (increased γH2AX and cleaved caspase 3, respectively) associated with significant reductions in proliferation (Ki67), ß6 expression and PDAC tumour growth. Conclusions: The FMDV-peptide drug conjugate SG3299 showed αvß6-selectivity in vitro and in vivo and can specifically eliminate αvß6-positive cancers, providing a promising new molecular- specific therapy for pancreatic cancer.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Dano ao DNA/efeitos dos fármacos , Integrinas/antagonistas & inibidores , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Antígenos de Neoplasias , Benzodiazepinas/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Knockout , Peptídeos/uso terapêutico , Pirróis/uso terapêutico
4.
Neurol Res ; 40(12): 1028-1039, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30213244

RESUMO

OBJECTIVE: Present study was to observe the neuroprotective ability of TMV coat protein by observing both in vitro studies on Rat PC-12 cells and in silico studies with LRRK2 receptor by molecular docking. METHODS: TMV coat protein was extracted out from the stem of Nicotiana tabacum and was purified and identified by MALDI-TOF/MS/MS analysis. We confer antioxidant activity of TMV coat protein by enzyme activity like superoxide dismutase (SOD), catalase (CAT) and glutathione-s-transferase (GST) and nonenzyme content by glutathione (GSH) and malondialdehyde (MDA) content. Neuroprotective ability of TMV coat protein was observed by determining the enzyme activity and nonenzyme content in treated cells that were exposed to neurotoxic shock. In silico studies were done in order to observe the molecular docking studies against LRRK2 receptor. RESULTS: Antioxidant content was found to be high in TMV coat protein and in treated Rat PC-12 cells as well. Enzyme activity and nonenzyme content were determined and their levels were found to be in increasing level with respect to the volume of 0.2 mg/ml of TMV coat protein. In silico studies revealed the binding efficacy of TMV coat protein with LRRK2 receptor by observing the molecular docking using automated servers. CONCLUSION: From the present study, it was found that TMV coat protein can be utilized as neuroprotective agent and inhibitor of LRRK2 receptor.


Assuntos
Proteínas do Capsídeo/uso terapêutico , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Simulação de Acoplamento Molecular , Fármacos Neuroprotetores/uso terapêutico , Animais , Proteínas do Capsídeo/química , Catalase/metabolismo , Fenômenos Químicos , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Malondialdeído , Fármacos Neuroprotetores/química , Células PC12/efeitos dos fármacos , Ratos , Superóxido Dismutase/metabolismo , Espectrometria de Massas em Tandem
5.
Nano Lett ; 18(5): 3007-3016, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29694053

RESUMO

Nanoparticulate vaccines can potentiate immune responses by site-specific drainage to lymph nodes (LNs). This approach may benefit from a nanoparticle engineering method with fine control over size and codelivery of antigen and adjuvant. Here, we applied the flash nanocomplexation (FNC) method to prepare nanovaccines via polyelectrolyte complexation of chitosan and heparin to coencapsulate the VP1 protein antigen from enterovirus 71, which causes hand-foot-mouth disease (HFMD), with tumor necrosis factor α (TNF) or CpG as adjuvants. FNC allows for reduction of the nanovaccine size to range from 90 to 130 nm with relatively narrower size distribution and a high payload capacity. These nanovaccines reached both proximal and distal LNs via subcutaneous injection and subsequently exhibited prolonged retention in the LNs. The codelivery induced strong immune activation toward a Th1 response in addition to a potent Th2 response, and conferred effective protection against lethal virus challenge comparable to that of an approved inactivated viral vaccine in mouse models of both passive and active immunization setting. In addition, these nanovaccines also elicited strong IgA titers, which may offer unique advantages for mucosal protection. This study addresses the issues of size control, antigen bioactivity retention, and biomanufacturing to demonstrate the translational potential of a subunit nanovaccine design.


Assuntos
Proteínas do Capsídeo/administração & dosagem , Sistemas de Liberação de Medicamentos , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Nanopartículas/química , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas Virais/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Animais , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/uso terapêutico , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/virologia , Humanos , Linfonodos/imunologia , Linfonodos/virologia , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Linfócitos T/virologia , Vacinas de Subunidades Antigênicas/uso terapêutico , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico
6.
J Allergy Clin Immunol ; 142(6): 1808-1817.e3, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29522849

RESUMO

BACKGROUND: Altered epithelial physical and functional barrier properties along with TH1/TH2 immune dysregulation are features of allergic asthma. Regulation of junction proteins to improve barrier function of airway epithelial cells has the potential for alleviation of allergic airway inflammation. OBJECTIVE: We sought to determine the immunomodulatory effect of knob protein of the adenoviral capsid on allergic asthma and to investigate its mechanism of action on airway epithelial junction proteins and barrier function. METHODS: Airway inflammation, including junction protein expression, was evaluated in allergen-challenged mice with and without treatment with knob. Human bronchial epithelial cells were exposed to knob, and its effects on expression of junction proteins and barrier integrity were determined. RESULTS: Administration of knob to allergen-challenged mice suppressed airway inflammation (eosinophilia, airway hyperresponsiveness, and IL-5 levels) and prevented allergen-induced loss of airway epithelial occludin and E-cadherin expression. Additionally, knob decreased expression of TH2-promoting inflammatory mediators, specifically IL-33, by murine lung epithelial cells. At a cellular level, treatment of human bronchial epithelial cells with knob activated c-Jun N-terminal kinase, increased expression of occludin and E-cadherin, and enhanced epithelial barrier integrity. CONCLUSION: Increased expression of junction proteins mediated by knob leading to enhanced epithelial barrier function might mitigate the allergen-induced airway inflammatory response, including asthma.


Assuntos
Proteínas do Capsídeo/farmacologia , Proteínas do Capsídeo/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Adenoviridae , Idoso , Animais , Brônquios/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Caderinas/metabolismo , Linhagem Celular , Citocinas/imunologia , Eosinofilia/imunologia , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Ocludina/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/imunologia
7.
Hum Gene Ther ; 28(11): 1061-1074, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28835127

RESUMO

Over the past decade, vectors derived from adeno-associated virus (AAV) have established themselves as a powerful tool for in vivo gene transfer, allowing long-lasting and safe transgene expression in a variety of human tissues. Nevertheless, clinical trials demonstrated how B and T cell immune responses directed against the AAV capsid, likely arising after natural infection with wild-type AAV, might potentially impact gene transfer safety and efficacy in patients. Seroprevalence studies have evidenced that most individuals carry anti-AAV neutralizing antibodies that can inhibit recombinant AAV transduction of target cells following in vivo administration of vector particles. Likewise, liver- and muscle-directed clinical trials have shown that capsid-reactive memory CD8+ T cells could be reactivated and expanded upon presentation of capsid-derived antigens on transduced cells, potentially leading to loss of transgene expression and immune-mediated toxicities. In celebration of the 25th anniversary of the European Society of Gene and Cell Therapy, this review article summarizes progress made during the past decade in understanding and modulating AAV vector immunogenicity. While the knowledge generated has contributed to yield impressive clinical results, several important questions remain unanswered, making the study of immune responses to AAV a priority for the field of in vivo transfer.


Assuntos
Dependovirus/genética , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Imunidade Celular/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/efeitos adversos , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/uso terapêutico , Ensaios Clínicos como Assunto , Dependovirus/imunologia , Terapia Genética/tendências , Vetores Genéticos/uso terapêutico , Humanos
8.
Hum Gene Ther ; 28(4): 338-349, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28323492

RESUMO

Recombinant adeno-associated viruses (AAVs) are quickly becoming the preferred viral vector for viral gene delivery for the treatment of a wide variety of genetic disorders. However, since their use in a clinical trial targeting hemophilia B patients 10 years ago, immune responses to the AAV capsid appear to have hampered some of the early clinical gene transfer efficacy. Indeed, AAV-based gene transfer has been shown to reactivate capsid-specific memory T cells, which have correlated with a decline in AAV-transduced tissue in some patients. Importantly, clinical trials have also shown that this reactivation can be quelled by administering time-course taper of glucocorticoid steroids before or after dosing. More recently, two clinical studies have shown that AAV gene transfer is not only able to induce a deleterious immune response, but also can result in the initiation of a tolerance to the AAV capsid mediated by regulatory T cells and exhausted T cells. This article reviews clinical trials describing immune responses to AAV, as well as the mechanisms responsible for immune tolerance in chronic infections and how it could apply to AAV-based gene transfer. A better understanding of both cytotoxic and tolerogenic immune responses to recombinant AAV will lead to safer gene transfer protocols in patients.


Assuntos
Proteínas do Capsídeo/uso terapêutico , Dependovirus/genética , Terapia Genética/efeitos adversos , Imunidade Celular , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos/efeitos adversos , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , Humanos , Linfócitos T/imunologia
9.
Biochem Biophys Res Commun ; 482(3): 506-513, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28212737

RESUMO

Glioblastoma (GBM) is the most common malignant brain tumor in adults. We designed an adeno-associated virus (AAV) vector for intracranial delivery of the secreted HSP70-targeted peptide APOPTIN derived from Apoptin to GBM tumors. We applied this therapy to GBM models using human U87MG glioma cells and GBM xenograft models in mice. In U87MG and U251MG cells, conditioned medium from AAV2-apoptin-derived peptide (ADP)-expressing cells induced 83% and 78% cell death. In mice bearing intracranial U87MG tumors treated with AAV2-ADP, treatment resulted in a significant decrease in tumor growth and longer survival in mice bearing orthotopic invasive GBM brain tumors. These data indicate that ssAAV2-ADP injection in the left hemisphere effectively prevented ipsilateral tumor growth but was insufficient to prevent distal tumor growth in the contralateral hemisphere. However, the systemic route is the most effective approach for treating widely dispersed tumors. In summary, systemic delivery of AAV2-ADP is an attractive approach for invasive GBM treatment.


Assuntos
Neoplasias Encefálicas/terapia , Proteínas do Capsídeo/uso terapêutico , Glioblastoma/terapia , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Dependovirus/genética , Feminino , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Glioblastoma/genética , Glioblastoma/patologia , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/genética , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Hum Gene Ther ; 28(4): 328-337, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28042943

RESUMO

Recombinant adenovirus-associated (rAAV) vectors due to their ease of construction, wide tissue tropism, and lack of pathogenicity remain at the forefront for long-term gene replacement therapy. In spite of very encouraging preclinical results, clinical trials were initially unsuccessful; expression of the rAAV vector-delivered therapeutic protein was transient. Loss of expression was linked to an expansion of AAV capsid-specific T-cell responses, leading to the hypothesis that rAAV vectors recall pre-existing memory T cells that had been induced by natural infections with AAV together with a helper virus. Although this was hotly debated at first, AAV capsid-specific T-cell responses were observed in several gene transfer trials that used high doses of rAAV vectors. Subsequent trials designed to circumvent these T-cell responses through the use of immunosuppressive drugs, rAAV vectors based on rare serotypes, or modified to allow for therapeutic levels of the transgene product at low, non-immunogenic vector doses are now successful in correcting debilitating diseases.


Assuntos
Proteínas do Capsídeo/uso terapêutico , Dependovirus/genética , Terapia Genética , Vetores Genéticos/uso terapêutico , Proteínas do Capsídeo/imunologia , Dependovirus/imunologia , Técnicas de Transferência de Genes/efeitos adversos , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/imunologia , Humanos , Linfócitos T/imunologia
11.
Eur Rev Med Pharmacol Sci ; 20(10): 2045-51, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27249603

RESUMO

The varied therapeutic approaches like radiotherapy, chemotherapy, surgery, etc. primarily aimed to target cancer cells specifically. Despite these efforts, they are not completely successful in eliminating this deadly pathological state. These failures ultimately lead to cancer reoccurrence, which is again, another burning problem associated with cancer. The prime reason for the above observation has been found to be the development of resistance by cancer cells towards cancer drugs or cancer-initiating cells (cancer stem cells) remain unaffected by existing treatment procedures. Recent research has evolved two drugs, salinomycin and apoptin, that hold great potential for the future of cancer treatment not only for restricting malignancy, but also in preventing tumor recurrence. The present review article will put light on these new upcoming cancer stem cell targeting agents.


Assuntos
Antineoplásicos/uso terapêutico , Células-Tronco Neoplásicas , Proteínas do Capsídeo/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Piranos/uso terapêutico
12.
Int J Pharm ; 502(1-2): 249-57, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-26915810

RESUMO

Virus-like particles (VLPs) have been exploited for various biomedical applications, such as the monitoring, prevention, diagnosis and therapy of disease. In this study, a novel multifunctional VLPs nanocarrier (TK-VLPs) was prepared and used for tumor-targeted delivery. The SPR and cell uptake results indicated that the TK peptide is a "bi-functional ligand" with high affinity for Caco-2, HRT-18 and HUVEC cells through the integrin α6ß1 and integrin αvß3 receptors. The results of the direct immunofluorescence, SDS-PAGE and western blot assays demonstrated that the TK-VLPs were successfully prepared using the baculovirus expression system. Confocal laser scanning microscopy and the flow cytometry analysis validated that the TK-VLPs could target to Caco-2, HRT-18 and HUVEC cells. An in vivo study further confirmed that the TK-VLPs could target and efficiently deliver fluorescein to tumor cells and the tumor vasculature in mice bearing subcutaneous tumors. TK-VLPs-DOX displayed a uniform, spherical shape and an average size of approximately 28nm. The results of the cell uptake and cytotoxicity assays indicated that TK-VLPs-DOX could enhance the selectivity for colorectal cancer cells. Together, our studies provide strong evidence that TK-VLPs could target colon tumor cells and tumor angiogenesis with enhanced permeability and retention effects, suggesting that the TK-VLPs are a multifunctional nanocarrier with potential applications in a colon tumor-targeted drug delivery system.


Assuntos
Antineoplásicos/administração & dosagem , Proteínas do Capsídeo/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Peptídeos/administração & dosagem , Timidina Quinase/química , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Baculoviridae/genética , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Peptídeos/química , Peptídeos/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico
13.
Eur Rev Med Pharmacol Sci ; 19(21): 4064-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26592827

RESUMO

Major current cancer strategies like surgery, radiotherapy, and chemotherapy are compromised due to major problem of recurrence, which usually lead to mortality. The widely accepted reason for this is resistance offered by cancer cells towards cancer drugs or inability of a therapeutic procedure to target real culprits viz. cancer-initiating cells (cancer stem cells). So, there is a current need of development of new agents targeting these cancer stem cells in order to overcome resistance to therapeutic procedures. The present review article is focused on new cancer cell targeting agents like salinomycin, apopotin etc and their mechanisms to target cancer stems cells will be discussed.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas do Capsídeo/uso terapêutico , Neoplasias/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Piranos/uso terapêutico , Animais , Antineoplásicos/farmacologia , Proteínas do Capsídeo/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular/métodos , Piranos/farmacologia
14.
Hum Gene Ther ; 26(10): 688-97, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26134901

RESUMO

Scalable production of recombinant adeno-associated virus vectors (rAAV) in baculovirus-infected Sf9 cells yields high burst sizes but variable infectivity rates per packaged AAV vector genome depending on the chosen serotype. Infectivity rates are particularly low for rAAV5 vectors, based on the genetically most divergent AAV serotype. In this study we describe key improvements of the OneBac system for the generation of rAAV5 vectors, whose manufacturing has been unsatisfactory in all current insect cell-based production systems. The Sf9 cell-based expression strategy for AAV5 capsid proteins was modified to enhance relative AAV5 VP1 levels. This resulted in a 100-fold boost of infectivity per genomic AAV5 particle with undiminished burst sizes per producer cell. Furthermore, the issue of collateral packaging of helper DNA into AAV capsids was approached. By modifications of the AAV rep and cap expression constructs used for the generation of stable Sf9 cell lines, collateral packaging of helper DNA sequences during rAAV vector production was dramatically reduced down to 0.001% of packaged rAAV genomes, while AAV5 burst sizes and infectivity rates were maintained. OneBac 2.0 represents the first insect cell-based scalable production system for high per-particle AAV5 infectivity rates combined with minimal collateral packaging of helper DNA, allowing the manufacturing of safe AAV5-based gene therapies for clinical application.


Assuntos
Proteínas do Capsídeo/genética , Dependovirus/genética , Terapia Genética , Vetores Genéticos , Baculoviridae/genética , Proteínas do Capsídeo/uso terapêutico , Humanos
15.
Indian J Exp Biol ; 53(5): 249-55, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26040021

RESUMO

Viral gene oncotherapy, targeted killing of cancer cells by viral genes, is an emerging non-infectious therapeutic cancer treatment modality. Chemo and radiotherapy in cancer treatment is limited due to their genotoxic side effects on healthy cells and need of functional p53, which is mutated in most of the cancers. VP3 (apoptin) of chicken infectious anaemia (CIA) and NS1 (Non structural protein 1) of Canine Parvovirus-2 (CPV-2) have been proven to have oncolytic potential in our laboratory. To evaluate oncolytic potential of VP3 and NS1 together these genes needed to be cloned in a bicistronic vector. In this study, both these genes were cloned and characterized for expression of their gene products and its apoptotic potential. The expression of VP3 and NS1 was studied by confocal microscopy and flowcytometry. Expression of VP3 and NS1 in pVIVO.VP3.NS1 transfected HeLa cells in comparison to mock transfected cells indicated that the double gene construct expresses both the products. This was further confirmed by flowcytometry where there was increase in cells expressing VP3 and NS1 in pVIVO.VP3.NS1 transfected group in comparison with the mock control group. The apoptotic inducing potential of this characterized pVIVO.VP3.NS1 was evaluated in human cervical cancer cell line (HeLa) by DNA fragmentation assay, TUNEL assay and Hoechst staning. This double construct was observed to induce apoptosis in HeLa cells.


Assuntos
Apoptose/genética , Proteínas do Capsídeo/genética , Vírus Oncogênicos/genética , Terapia Viral Oncolítica , Proteínas não Estruturais Virais/genética , Animais , Proteínas do Capsídeo/uso terapêutico , Galinhas/virologia , Cães , Vetores Genéticos , Células HeLa , Humanos , Parvovirus Canino/genética , Proteínas não Estruturais Virais/uso terapêutico
16.
PLoS One ; 10(5): e0125851, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25933160

RESUMO

One strategy in cancer immunotherapy is to capitalize on the key immunoregulatory and antigen presenting capabilities of dendritic cells (DCs). This approach is dependent on efficient delivery of tumor specific antigens to DCs, which subsequently induce an anti-tumor T-cell mediated immune response. Human adenovirus serotype 5 (HAdV5) has been used in human studies for gene delivery, but has limited infection in DCs, which lack the proper receptors. Addition of the porcine fiber knob (PK) from porcine adenovirus type 4 to HAdV5 allows the virus to deliver genetic material via binding to glycosylated surface proteins and bypasses the coxsackie-and-adenovirus receptor required by wild-type HAdV5. In this study we explored the potential therapeutic applications of an adenovirus with PK-based tropism against cancers expressing mesothelin. Infectivity and gene transfer assays were used to compare Ad5-PK to wild-type HAdV5. Mouse models were used to demonstrate peptide specificity and T-cell responses. We show that the PK modification highly augmented infection of DCs, including the CD141+ DC subset, a key subset for activation of naïve CD8+ T-cells. We also show that Ad5-PK increases DC infectivity and tumor specific antigen expression. Finally, vaccination of mice with the Ad5-PK vector resulted in enhanced T-cell-mediated interferon gamma (IFN-γ) release in response to both mesothelin peptide and a tumor line expressing mesothelin. Ad5-PK is a promising tool for cancer immunotherapy as it improves infectivity, gene transfer, protein expression, and subsequent T-cell activation in DCs compared to wild-type HAdV5 viruses.


Assuntos
Proteínas do Capsídeo/uso terapêutico , Terapia Genética , Imunoterapia , Neoplasias/terapia , Adenovírus Humanos , Adenovirus Suínos/genética , Adenovirus Suínos/imunologia , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Células Dendríticas/imunologia , Proteínas Ligadas por GPI/biossíntese , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Mesotelina , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Suínos , Transdução Genética
17.
ACS Appl Mater Interfaces ; 7(22): 12089-98, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-25989427

RESUMO

Here we explore the formation of virus-like nanoparticles (VNPs) utilizing 22-24 nm iron oxide nanoparticles (NPs) as cores and proteins derived from viral capsids of brome mosaic virus (BMV) or hepatitis B virus (HBV) as shells. To accomplish that, hydrophobic FeO/Fe3O4 NPs prepared by thermal decomposition of iron oleate were coated with poly(maleic acid-alt-octadecene) modified with poly(ethylene glycol) (PEG) tails of different lengths and grafting densities. MRI studies show high r2/r1 relaxivity ratios of these NPs that are practically independent of the polymer coating type. The versatility and flexibility of the viral capsid protein are on display as they readily form shells that exceed their native size. The location of the long PEG tails upon shell formation was investigated by electron microscopy and small-angle X-ray scattering. PEG tails were located differently in the BMV and HBV VNPs, with the BMV VNPs preferentially entrapping the tails in the interior and the HBV VNPs allowing the tails to extend through the capsid, which highlights the differences between intersubunit interactions in these two icosahedral viruses. The robustness of the assembly reaction and the protruding PEG tails, potentially useful in modulating the immune response, make the systems introduced here a promising platform for biomedical applications.


Assuntos
Bromovirus/química , Proteínas do Capsídeo/química , Vírus da Hepatite B/química , Nanopartículas/química , Proteínas do Capsídeo/uso terapêutico , Compostos Férricos/química , Compostos Férricos/uso terapêutico , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imageamento por Ressonância Magnética , Nanopartículas/uso terapêutico , Polietilenoglicóis/química , Polietilenoglicóis/uso terapêutico , Polímeros/química , Polímeros/uso terapêutico
18.
Sci Rep ; 4: 6808, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25348392

RESUMO

Nanomaterials with near-infrared (NIR) absorption have been widely studied in cancer detection and photothermal therapy (PTT), while it remains a great challenge in targeting tumor efficiently with minimal side effects. Herein we report a novel multifunctional phage-mimetic nanostructure, which was prepared by layer-by-layer self-assembly of Au@Ag heterogenous nanorods (NRs) with rhodamine 6G, and specific pVIII fusion proteins. Au@Ag NRs, first being applied for PTT, exhibited excellent stability, cost-effectivity, biocompatibility and tunable NIR absorption. The fusion proteins were isolated from phage DDAGNRQP specifically selected from f8/8 landscape phage library against colorectal cancer cells in a high-throughput way. Considering the definite charge distribution and low molecular weight, phage fusion proteins were assembled on the negatively charged NR core by electrostatic interactions, exposing the N-terminus fused with DDAGNRQP peptide on the surface. The fluorescent images showed that assembled phage fusion proteins can direct the nanostructure into cancer cells. The nanostructure was more efficient than gold nanorods and silver nanotriangle-based photothermal agents and was capable of specifically ablating SW620 cells after 10 min illumination with an 808 nm laser in the light intensity of 4 W/cm(2). The prepared nanostructure would become an ideal reagent for simutaneously targeted optical imaging and PTT of tumor.


Assuntos
Biomimética , Proteínas do Capsídeo/química , Neoplasias Colorretais/terapia , Nanoestruturas/química , Fototerapia , Proteínas do Capsídeo/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Colorretais/ultraestrutura , Neoplasias Colorretais/virologia , Ouro/química , Humanos , Nanoestruturas/uso terapêutico , Nanotubos/química , Rodaminas/química , Rodaminas/uso terapêutico , Prata/química , Espectroscopia de Luz Próxima ao Infravermelho
19.
Adv Exp Med Biol ; 818: 11-37, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25001530

RESUMO

The virus-derived protein Apoptin has the ability to induce p53-independent apoptosis in a variety of human cancer cells while leaving normal cells unharmed. It thus represents a potential anti-cancer therapeutic agent of the future but a proper understanding of Apoptin-induced signalling events is necessary prior to clinical application. The tumor-specific nuclear translocation and phosphorylation of Apoptin by a cellular kinase such as protein kinase C seem to be required for its function but otherwise the mode of tumor selectivity remains unknown. Apoptin has been shown to interact with several cellular proteins including Akt and the anaphase-promoting complex that regulate its activity and promote caspase-dependent apoptosis. This chapter summarizes the available data on tumor-specific pathways sensed by Apoptin and the mechanism of Apoptin-induced cell death.


Assuntos
Antineoplásicos/metabolismo , Proteínas do Capsídeo/metabolismo , Núcleo Celular/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Transporte Ativo do Núcleo Celular , Animais , Antineoplásicos/uso terapêutico , Apoptose , Proteínas do Capsídeo/uso terapêutico , Caspases/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Proteína Quinase C/metabolismo , Proteína Supressora de Tumor p53/metabolismo
20.
Spinal Cord ; 52(1): 3-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24190077

RESUMO

OBJECTIVE: The objective of this study is to evaluate the safety and efficacy of a tumor-specific apoptosis-inducing gene, apoptin, as delivered by the non-viral carrier, PAM-RG4, in an animal model of spinal cord tumor. METHODS: Male Sprague-Dawley rats were given a 2.5-µl intramedullary injection of C6 glioma (100,000) cells and randomized into three groups (day 0). On day 5, animals received a 7.5-µl intramedullary injection of Dulbecco's modified Eagle's medium (Group 1; n=7), PAM-RG4/control gene polyplex (Group 2; n=7), or PAM-RG4/apoptin gene polyplex (Group 3; n=8). Hindlimb functional strength was assessed every other day for the duration of the study. The spinal cords of killed animals were collected and hematoxylin-eosin stained. RESULTS: Following treatment, animals that received apoptin had significantly higher mean functional hindlimb scores than those of sham control animals, showing a level of preserved hindlimb function throughout the study. In addition, Group 1 (sham control) and Group 2 (control gene) animals had median survival scores lower than those of animals receiving apoptin. Histopathological analysis showed marked retardation of tumor progression in apoptin-treated animals compared with sham controls. CONCLUSION: Our study suggests that apoptin is safe for use in the mammalian spinal cord as well as effective in slowing the progression of tumor growth in the spinal cord. The significant slowing of tumor progression, as manifested by the preserved hindlimb function, coupled with the reduction in tumor volume, shows local non-viral delivery of apoptin could serve as an emerging therapy for the treatment of intramedullary spinal cord tumors.


Assuntos
Proteínas do Capsídeo/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Paresia/prevenção & controle , Neoplasias da Medula Espinal/complicações , Animais , Proteínas do Capsídeo/uso terapêutico , Modelos Animais de Doenças , Masculino , Paresia/etiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA