Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Immunology ; 172(3): 375-391, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38471664

RESUMO

Persistent human papillomavirus (HPV) infection is associated with multiple malignancies. Developing therapeutic vaccines to eliminate HPV-infected and malignant cells holds significant value. In this study, we introduced a lipid nanoparticle encapsulated mRNA vaccine expressing tHA-mE7-mE6. Mutations were introduced into E6 and E7 of HPV to eliminate their tumourigenicity. A truncated influenza haemagglutinin protein (tHA), which binds to the CD209 receptor on the surface of dendritic cells (DCs), was fused with mE7-mE6 in order to allow efficient uptake of antigen by antigen presenting cells. The tHA-mE7-mE6 (mRNA) showed higher therapeutic efficacy than mE7-mE6 (mRNA) in an E6 and E7+ tumour model. The treatment resulted in complete tumour regression and prevented tumour formation. Strong CD8+ T-cell immune response was induced, contributing to preventing and curing of E6 and E7+ tumour. Antigen-specific CD8+ T were found in spleens, peripheral blood and in tumours. In addition, the tumour infiltration of DC and NK cells were increased post therapy. In conclusion, this study described a therapeutic mRNA vaccine inducing strong anti-tumour immunity in peripheral and in tumour microenvironment, holding promising potential to treat HPV-induced cancer and to prevent cancer recurrence.


Assuntos
Vacinas Anticâncer , Células Dendríticas , Proteínas Oncogênicas Virais , Proteínas E7 de Papillomavirus , Infecções por Papillomavirus , Vacinas contra Papillomavirus , Vacinas de mRNA , Animais , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/prevenção & controle , Proteínas E7 de Papillomavirus/imunologia , Vacinas Anticâncer/imunologia , Proteínas Oncogênicas Virais/imunologia , Proteínas Oncogênicas Virais/genética , Vacinas contra Papillomavirus/imunologia , Células Dendríticas/imunologia , Humanos , Camundongos , Feminino , Linfócitos T CD8-Positivos/imunologia , Camundongos Endogâmicos C57BL , Nanopartículas , Células Apresentadoras de Antígenos/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Células Matadoras Naturais/imunologia , Proteínas Repressoras/imunologia , Proteínas Repressoras/genética , Neoplasias/terapia , Neoplasias/imunologia , RNA Mensageiro/genética , Linhagem Celular Tumoral , Lipossomos
2.
Prostate ; 78(6): 457-468, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29450905

RESUMO

BACKGROUND: Patients with advanced prostate cancer have limited curative options, therefore new treatments are needed. Mouse models play a pivotal role in the discovery and development of new treatments. In the present study, a TRAMP-derived Orthotopic Prostate Syngeneic (TOPS) mouse model was developed and found to provide a consistent means of monitoring tumor and metastatic responses to novel treatments. METHODS: The mouse TOPS model was generated using luciferase transduced TRAMP-C2 prostate cancer cells that were orthotopically injected into Bl6 mice by ultrasound guidance. Tumor growth and development was monitored using ultrasound and bioluminescence imaging. RESULTS: Tumors and metastases were consistently established and increases in tumor size correlated with increases in bioluminescence. In addition, when mice with an established tumor were castrated, tumor progression mirrored clinical progression. We further treated the TOPS model with an oncolytic Herpes Simplex virus and showed that we were able to monitor the therapeutic effect of the orthotopic tumor after virus treatment through IVIS imaging system. CONCLUSION: We have developed a powerful animal model to advance the current selection of effective treatments for patients with advanced prostate cancer.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Adenocarcinoma/tratamento farmacológico , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico
3.
Biochem Biophys Res Commun ; 497(1): 51-57, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29421661

RESUMO

Transactive response DNA-binding protein 43kD (TDP-43) is a major component of tau-negative and ubiquitin-positive inclusions that characterize ALS (amyotrophic lateral sclerosis) and FTLD (frontotemporal lobar degeneration). Due to its central role in neurodegenerative disease pathogenesis, most research recently has focused on its role associated with neurodegeneration disease, research on neuron and glial cell showed that pathological TDP-43 is associated with cell apoptosis which lead to loss of functional neurons and glial cells. However, little is known about its role on cancer cells, here we report a 35kD fragment of TDP-43 also plays a key role in apoptosis of breast cancer cells, and may be served as a potential therapeutic target to cure cancer.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3/metabolismo , Proteínas de Ligação a DNA/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Humanos , Células MCF-7
4.
Blood ; 127(21): 2575-86, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-26941401

RESUMO

Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML.


Assuntos
Herpesvirus Humano 1/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Leucemia/imunologia , Raios Ultravioleta , Inativação de Vírus/efeitos da radiação , Degranulação Celular/imunologia , Movimento Celular/imunologia , Feminino , Humanos , Interleucina-15/imunologia , Interleucina-2/imunologia , Células Jurkat , Masculino , NF-kappa B/imunologia , Proteína Quinase C/imunologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia
5.
J Cell Physiol ; 231(6): 1350-63, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26529564

RESUMO

It has long been realized that hematopoietic cells may have the capacity to trans-differentiate into non-lymphohematopoietic cells under specific conditions. However, the mechanisms and the factors for hematopoietic cell trans-differentiation remain unknown. In an in vitro culture system, we found that using a conditioned medium from proliferating fibroblasts can induce a subset of hematopoietic cells to become adherent fibroblast-like cells (FLCs). FLCs are not fibroblasts nor other mesenchymal stromal cells, based on their expression of type-1 collagen, and other stromal cell marker genes. To identify the active factors in the conditioned medium, we cultured fibroblasts in a serum-free medium and collected it for further purification. Using the fractions from filter devices of different molecular weight cut-offs, and ammonium sulfate precipitation collected from the medium, we found the active fraction is a protein. We then purified this fraction by using fast protein liquid chromatography (FPLC) and identified it by mass spectrometer as macrophage colony-stimulating factor (M-CSF). The mechanisms of M-CSF-inducing trans-differentiation of hematopoietic cells seem to involve a tyrosine kinase signalling pathway and its known receptor. The FLCs express a number of stem cell markers including SSEA-1 and -3, OCT3/4, NANOG, and SOX2. Spontaneous and induced differentiation experiments confirmed that FLCs can be further differentiated into cell types of three germ layers. These data indicate that hematopoietic cells can be induced by M-CSF to dedifferentiate to multipotent stem cells. This study also provides a simple method to generate multipotent stem cells for clinical applications.


Assuntos
Tecido Adiposo/metabolismo , Transdiferenciação Celular , Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucócitos Mononucleares/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Comunicação Parácrina , Baço/metabolismo , Adipócitos/metabolismo , Adipogenia , Tecido Adiposo/citologia , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Fenótipo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais , Baço/citologia
6.
Mol Ther ; 21(4): 842-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23337981

RESUMO

Vesicular stomatitis virus (VSV) is an oncolytic virus which selectively infects and kills cancer cells. The goal of the present study was to determine whether VSV is capable of targeting metastatic lesions that arise in situ in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. The interferon (IFN)-responsive luciferase containing VSV(AV3) strain was injected intraprostatically into both control and TRAMP mice. Distribution, infectivity, apoptosis, and status of the IFN response were evaluated at the site of viral injection (prostate), as well as in metastatic lesions (lymph nodes), through plaque, polymerase chain reaction (PCR), and immunohistochemical analysis. Bioluminescence analyses demonstrated that VSV(AV3) persisted at high levels in the prostate region of TRAMP mice for up to 96 hours, but at relatively low levels and for only 48 hours in control mice. Live virus was discovered in the lymph nodes of TRAMP mice, but not in control mice. TUNEL staining revealed increased cell death in VSV(AV3) infected metastatic cells present in the lymph nodes of TRAMP mice. There was an evidence of IFN activation in lymph nodes containing metastatic cells. Our results indicate that intraprostatic injections of VSV(AV3) can be used as a means to infect and kill metastatic lesions associated with advanced prostate cancer.


Assuntos
Adenocarcinoma/terapia , Metástase Neoplásica/prevenção & controle , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Vírus da Estomatite Vesicular Indiana/genética
7.
Biopharm Drug Dispos ; 35(2): 104-18, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24151189

RESUMO

20(S)-Protopanaxadiol (aPPD), a ginseng sapogenin, has been shown to be a promising anti-cancer compound and anti-depressant agent. Although the bacterial biotransformation of ginsenosides has been studied thoroughly, few have reported on the cytochrome P450 (P450) mediated metabolism of aPPD. Taken orally, aPPD must first undergo absorption and metabolism in the intestine before further metabolism in the liver. The present study investigated the comparative biotransformation profile of aPPD in human intestinal microsomes (HIM) and human liver microsomes (HLM) and characterized the human P450 enzymes involved in aPPD metabolism. Three major monooxygenated metabolites and five minor dioxygenated metabolites were identified as the predominant products in aPPD incubations with HIM and HLM using liquid chromatography-mass spectrometry. Reaction phenotyping studies were performed with a panel of specific P450 chemical inhibitors, antibody inhibition and human recombinant P450 enzymes. Ketoconazole, a CYP3A inhibitor, blocked the formation of oxygenated metabolites of aPPD in both HIM and HLM in a concentration dependent manner. Among the human recombinant P450 enzymes assayed, CYP3A4 exhibited the highest activity towards aPPD oxidative metabolite formation, followed by CYP3A5. In summary, the results have shown that aPPD is extensively metabolized by HIM and the metabolite profile following in vitro incubations is similar in HIM and HLM. CYP3A4 and CYP3A5 isoforms are the predominant enzymes responsible for oxygenation of aPPD in HIM and HLM. The characterization of aPPD as a CYP3A substrate may facilitate better prediction of drug-herb interactions when aPPD is taken concomitantly with other therapeutic agents.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Microssomos/metabolismo , Sapogeninas/farmacocinética , Biotransformação , Inibidores das Enzimas do Citocromo P-450/farmacologia , Humanos , Isoenzimas/metabolismo
8.
Oncol Lett ; 27(6): 244, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38638849

RESUMO

Sarcoma is derived from mesenchymal neoplasms and has numerous subtypes, accounting for 1% of all adult malignancies and 15% of childhood malignancies. The prognosis of metastatic or recurrent sarcoma remains poor. The current study presents two cases of sarcoma enrolled in a phase I dose escalation trial for solid tumor, who had previously failed all standard therapies. These patients were treated with VG161, an immune-stimulating herpes simplex virus type 1 oncolytic virus with payloads of IL-12, IL-15 and IL-15 receptor α unit, and a programmed cell death 1 (PD-1)/PD-1 ligand 1 blocking peptide. Both cases demonstrated stable disease as the best response, accompanied by a noteworthy prolongation of progression-free survival (11.8 months for chondrosarcoma and 11.9 months for soft tissue sarcoma, respectively) at a dose of 2.5×108 PFU/cycle. In addition, the treatment led to the activation of anti-cancer immunity, as evident from cytokine, lymphocyte subset and related pathway analyses of peripheral blood and/or tumor biopsy samples. These promising results suggest that VG161 monotherapy holds promise as an effective treatment for sarcoma and warrants further investigation through clinical trials. The two reported patients were part of a phase I clinical trial conducted and registered on the Australian New Zealand Clinical Trials Registry in Australia (registration no. ACTRN12620000244909; registration date, 26 February, 2020).

9.
Virol J ; 10: 241, 2013 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-23876001

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality worldwide, and novel treatment modalities to improve the prognosis of patients with advanced disease are highly desirable. Oncolytic virotherapy is a promising approach for the treatment of advanced NSCLC. MicroRNAs (miRNAs) may be a factor in the regulation of tumor-specific viral replication. The purpose of this study was to investigate whether miRNA-145 regulated oncolytic herpes simplex virus-1 (HSV-1) can selectively kill NSCLC cells with reduced collateral damage to normal cells. METHODS: We incorporated 4 copies of miRNA-145 target sequences into the 3'-untranslated region of an HSV-1 essential viral gene, ICP27, to create AP27i145 amplicon viruses and tested their target specificity and toxicity on normal cells and lung cancer cells in vitro. RESULTS: miRNA-145 expression in normal cells was higher than that in NSCLC cells. AP27i145 replication was inversely correlated with the expression of miRNA-145 in infected cells. This oncolytic HSV-1 selectively reduced cell proliferation and prevented the colony formation of NSCLC cells. The combination of radiotherapy and AP27i145 infection was significantly more potent in killing cancer cells than each therapy alone. CONCLUSIONS: miRNA-145-regulated oncolytic HSV-1 is a promising agent for the treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , MicroRNAs/metabolismo , Vírus Oncolíticos/fisiologia , Regiões 3' não Traduzidas , Sítios de Ligação , Proliferação de Células , Sobrevivência Celular , DNA Viral/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Humanos , Vírus Oncolíticos/genética , Vírus Oncolíticos/crescimento & desenvolvimento
10.
Viruses ; 15(8)2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-37632102

RESUMO

RNA vaccines, including conventional messenger RNA (mRNA) vaccines, circular RNA (circRNA) vaccines, and self-amplifying RNA (saRNA) vaccines, have ushered in a promising future and revolutionized vaccine development. The success of mRNA vaccines in combating the COVID-19 pandemic caused by the SARS-CoV-2 virus that emerged in 2019 has highlighted the potential of RNA vaccines. These vaccines possess several advantages, such as high efficacy, adaptability, simplicity in antigen design, and the ability to induce both humoral and cellular immunity. They also offer rapid and cost-effective manufacturing, flexibility to target emerging or mutant pathogens and a potential approach for clearing immunotolerant microbes by targeting bacterial or parasitic survival mechanisms. The self-adjuvant effect of mRNA-lipid nanoparticle (LNP) formulations or circular RNA further enhances the potential of RNA vaccines. However, some challenges need to be addressed. These include the technology's immaturity, high research expenses, limited duration of antibody response, mRNA instability, low efficiency of circRNA cyclization, and the production of double-stranded RNA as a side product. These factors hinder the widespread adoption and utilization of RNA vaccines, particularly in developing countries. This review provides a comprehensive overview of mRNA, circRNA, and saRNA vaccines for infectious diseases while also discussing their development, current applications, and challenges.


Assuntos
COVID-19 , Vacina Antivariólica , Humanos , RNA Circular , Pandemias , COVID-19/prevenção & controle , SARS-CoV-2/genética , RNA Mensageiro , RNA de Cadeia Dupla
11.
Mol Ther Oncolytics ; 28: 334-348, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36938544

RESUMO

VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing virulence. The TTDR platform is based on transcriptional control of the essential HSV-1 immediate-early protein ICP27 using a tumor-specific carcinoembryonic antigen (CEA) promoter, coupled with translational control of the neurovirulence factor ICP34.5 using multiple microRNA (miR)-binding sites. VG2025 further incorporates IL-12 and the IL-15/IL-15 receptor alpha subunit complex to enhance the antitumor and immune stimulatory properties of oncolytic HSVs. The TTDR strategy was verified in vitro and shown to be highly selective. Strong in vivo antitumor efficacy was observed following both intratumoral and intravenous administration. Clear abscopal and immune memory effects were also evident, indicating a robust antitumor immune response. Gene expression profiling of treated tumors revealed increased immune cell infiltration and activation of multiple immune-signaling pathways when compared with the backbone virus. Absence of neurotoxicity was verified in mice and in rhesus monkeys. Taken together, the enhanced tumor clearance, excellent safety profile, and positive correlation between CEA levels and viral replication efficiency may provide an opportunity for using biomarker-based precision medicine in oncolytic virotherapy.

12.
Nat Commun ; 14(1): 5925, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37739969

RESUMO

The recent outbreaks of mpox have raised concerns over the need for effective vaccines. However, the current approved vaccines have either been associated with safety concerns or are in limited supply. mRNA vaccines, which have shown high efficacy and safety against SARS-CoV-2 infection, are a promising alternative. In this study, three mRNA vaccines are developed that encode monkeypox virus (MPXV) proteins A35R and M1R, including A35R extracellular domain -M1R fusions (VGPox 1 and VGPox 2) and a mixture of encapsulated full-length mRNAs for A35R and M1R (VGPox 3). All three vaccines induce early anti-A35R antibodies in female Balb/c mice, but only VGPox 1 and 2 generate detectable levels of anti-M1R antibodies at day 7 after vaccination. However, all three mRNA vaccine groups completely protect mice from a lethal dose of vaccinia virus (VACV) challenge. A single dose of VGPox 1, 2, and 3 provide protection against the lethal viral challenge within 7 days post-vaccination. Long-term immunity and protection were also observed in all three candidates. Additionally, VGPox 2 provided better passive protection. These results suggest that the VGPox series vaccines enhance immunogenicity and can be a viable alternative to current whole-virus vaccines to defend against mpox.


Assuntos
COVID-19 , Mpox , Feminino , Animais , Camundongos , Vaccinia virus/genética , Monkeypox virus/genética , SARS-CoV-2 , Camundongos Endogâmicos BALB C , Vacinas de mRNA
13.
Vaccines (Basel) ; 11(12)2023 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-38140209

RESUMO

The development of effective cancer vaccines remains a significant challenge due to immune tolerance and limited clinical benefits. Oncolytic herpes simplex virus type 1 (oHSV-1) has shown promise as a cancer therapy, but efficacy is often limited in advanced cancers. In this study, we constructed and characterized a novel oHSV-1 virus (VG22401) expressing the human epidermal growth factor receptor 2 (HER2), a transmembrane glycoprotein overexpressed in many carcinomas. VG22401 exhibited efficient replication and HER2 payload expression in both human and mouse colorectal cancer cells. Mice immunized with VG22401 showed significant binding of serum anti-HER2 antibodies to HER2-expressing tumor cells, inducing antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Furthermore, mice primed with VG22401 and intratumorally boosted with the same virus showed enhanced antitumor efficacy in a bilateral syngeneic HER2(+) tumor model, compared to HER2-null backbone virus. This effect was accompanied by the induction of anti-HER2 T cell responses. Our findings suggest that peripheral priming with HER2-expressing oHSV-1 followed by an intratumoral boost with the same virus can significantly enhance antitumor immunity and efficacy, presenting a promising strategy for cancer immunotherapy.

14.
Antiviral Res ; 212: 105556, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36871919

RESUMO

The coronavirus SARS-CoV-2 has mutated quickly and caused significant global damage. This study characterizes two mRNA vaccines ZSVG-02 (Delta) and ZSVG-02-O (Omicron BA.1), and associating heterologous prime-boost strategy following the prime of a most widely administrated inactivated whole-virus vaccine (BBIBP-CorV). The ZSVG-02-O induces neutralizing antibodies that effectively cross-react with Omicron subvariants. In naïve animals, ZSVG-02 or ZSVG-02-O induce humoral responses skewed to the vaccine's targeting strains, but cellular immune responses cross-react to all variants of concern (VOCs) tested. Following heterologous prime-boost regimes, animals present comparable neutralizing antibody levels and superior protection against Delta and Omicron BA.1variants. Single-boost only generated ancestral and omicron dual-responsive antibodies, probably by "recall" and "reshape" the prime immunity. New Omicron-specific antibody populations, however, appeared only following the second boost with ZSVG-02-O. Overall, our results support a heterologous boost with ZSVG-02-O, providing the best protection against current VOCs in inactivated virus vaccine-primed populations.


Assuntos
COVID-19 , Animais , Humanos , COVID-19/prevenção & controle , Vacinas contra COVID-19 , SARS-CoV-2/genética , Anticorpos Neutralizantes , Vacinas de mRNA , Anticorpos Antivirais , Vacinas de Produtos Inativados
15.
J Neurosci ; 31(31): 11126-32, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21813674

RESUMO

Frontotemporal dementia (FTD) has been linked to mutations in the progranulin gene (GRN) that lead to progranulin (PGRN) haploinsufficiency. Thus far, our understanding of the effects of PGRN depletion in the brain has been derived from investigation of gross pathology, and more detailed analyses of cellular function have been lacking. We report that knocking down PGRN levels in rat primary hippocampal cultures reduces neural connectivity by decreasing neuronal arborization and length as well as synapse density. Despite this, the number of synaptic vesicles per synapse and the frequency of mEPSCs are increased in PGRN knockdown cells, suggesting an increase in the probability of release at remaining synapses. Interestingly, we demonstrate that the number of vesicles per synapse is also increased in postmortem brain sections from FTD patients with PGRN haploinsufficiency, relative to controls. Our observations show that PGRN knockdown severely alters neuronal connectivity in vitro and that the synaptic vesicle phenotype observed in culture is consistent with that observed in the hippocampus of FTD patients.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Demência Frontotemporal/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Neurônios/fisiologia , Sinapses/fisiologia , Idoso , Análise de Variância , Animais , Células Cultivadas , Dendritos/ultraestrutura , Proteína 4 Homóloga a Disks-Large , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Demência Frontotemporal/genética , Proteínas de Fluorescência Verde/genética , Guanilato Quinases/genética , Hipocampo/citologia , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Luminescentes/genética , Masculino , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Mutação , Compostos de Piridínio/metabolismo , Compostos de Amônio Quaternário/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Receptores de AMPA/metabolismo , Sinapses/genética , Sinapses/ultraestrutura , Vesículas Sinápticas/genética , Vesículas Sinápticas/fisiologia , Vesículas Sinápticas/ultraestrutura , Sinaptofisina/metabolismo , Sais de Tetrazólio , Tiazóis , Transfecção/métodos
16.
Anticancer Drugs ; 23(5): 543-52, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22481061

RESUMO

This study focuses on determining the pharmacokinetics, biodistribution, and efficacy of the ginsenoside aglycone protopanaxadiol (aPPD) administered as a single agent in a novel oral dosage formulation. To obtain these data and to characterize the stability of aPPD, appropriate analytical assay development was carried out. The solubility and stability of aPPD were determined, and the compound was formulated for oral gavage. aPPD levels in blood and tissues following oral administration to nu/nu nude mice were determined using liquid chromatography-mass spectrometry/mass spectrometry. The efficacy of aPPD was determined upon oral administration to nu/nu nude mice bearing PC-3 human prostate cancer xenograft tumors. Immunohistochemical analysis of tumor tissues was performed to establish apoptotic indices and Ki-67 expression as markers of proliferation. The maximum solubility of aPPD in ethanol was 68.4 mg/ml. aPPD administered at a dose of 70 mg/kg yielded a T(max) of approximately 40 min and a C(max) value of 3.9 ± 1.4 µg/ml, and no toxicity was observed. aPPD accumulated largely in the stomach and small intestine and was also present in the brain. This dose engendered a significant delay in PC-3 tumor growth, an increase in apoptotic index, and a decrease in Ki-67 levels. We have shown that aPPD is a stable compound that can be formulated for oral gavage. Pharmacokinetic studies demonstrate the ability of this compound to be absorbed after oral administration. Future studies will assess the activity and pharmacokinetics of aPPD when administered in combination with standard chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Ginsenosídeos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Sapogeninas/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Química Farmacêutica , Cromatografia Líquida de Alta Pressão , Composição de Medicamentos , Estabilidade de Medicamentos , Ginsenosídeos/administração & dosagem , Ginsenosídeos/farmacocinética , Ginsenosídeos/farmacologia , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Dose Máxima Tolerável , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias da Próstata/sangue , Neoplasias da Próstata/metabolismo , Sapogeninas/administração & dosagem , Sapogeninas/farmacocinética , Sapogeninas/farmacologia , Extração em Fase Sólida , Solubilidade , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Biol Rep ; 39(1): 343-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21556766

RESUMO

HIV-1 integrase (HIV-1 IN), a key element of HIV-1-derived lentiviral vectors, is crucial for the stable maintenance of the vector gene by inserting them into host genome. HIV-1 IN has been found to have functions other than integration, such as involving in virion morphology, viral DNA synthesis and viral DNA nuclear import. In our study, the yeast two-hybrid assay identified a tetrapeptide 156KELK159 in HIV-1 IN that was crucial for HIV-1 IN and Daxx interaction. To investigate the functions of the tetrapeptide 156KELK159 of the HIV-1 IN, both the wild type HIV-1 IN and a mutant without 156KELK159 were used to package the EGFP reporter gene contained lentivirus. p24 based titer assay revealed that deleting the tetrapeptide did not affect virus packaging. The result was verified by quantitative real time PCR with viral specific primers. But the 156KELK159 was crucial for lentiviral gene integration. Deleting the tetrapeptide made the percentage of cells expressing the reporter gene significantly decreased and did not affect the level of DNA entered into the cells or nucleus. Real time reverse transcription PCR and FACS were used to detect the lentiviral report gene expression in infection maintaining cells and revealed 156KELK159 did not affect lentiviral vector gene expression. Our results may shed light on the regulatory mechanism of gene integration of lentivirus.


Assuntos
Integrase de HIV/genética , HIV-1/genética , Oligopeptídeos/fisiologia , Transdução Genética/métodos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Proteínas Correpressoras , Primers do DNA/genética , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Oligopeptídeos/genética , Plasmídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido
18.
Mol Ther Oncolytics ; 27: 89-99, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36321136

RESUMO

Genetic modification of coxsackievirus B3 (CVB3) by inserting target sequences (TS) of tumor-suppressive and/or organ-selective microRNAs (miRs) into viral genome can efficiently eliminate viral pathogenesis without significant impacts on its oncolytic activity. Nonetheless, reversion mutants (loss of miR-TS inserts) were identified as early as day 35 post-injection in ∼40% immunodeficient mice. To improve the stability, here we re-engineered CVB3 by (1) replacing the same length of viral genome at the non-coding region with TS of cardiac-selective miR-1/miR-133 and pancreas-enriched miR-216/miR-375 or (2) inserting the above miR-TS into the coding region (i.e., P1 region) of viral genome. Serial passaging of these newly established miR-CVB3s in cultured cells for 20 rounds demonstrated significantly improved stability compared with the first-generation miR-CVB3 with 5'UTR insertion of miR-TS. The safety and stability of these new miR-CVB3s was verified in immunocompetent mice. Moreover, we showed that these new viruses retained the ability to suppress lung tumor growth in a xenograft mouse model. Finally, we observed that miR-CVB3 with insertion in P1 region was more stable than miR-CVB3 with preserved length of the 5'UTR, whereas the latter displayed significantly higher oncolytic activity. Overall, we presented here valid strategies to enhance the genomic stability of miR-CVB3 for virotherapy.

19.
Viruses ; 14(11)2022 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-36366425

RESUMO

Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 (HSV-1) or multicycle treatment with OVs on anticancer efficacy of VG161, an HSV-1 OV in phase 2 clinical trial. VG161 efficacy was tested in CT26 mouse models by comparing the efficacy and immune response in naïve mice or in mice that were immunized with VG161. Moreover, VG161 efficacy in HLA-matched CD34+ humanized intrahepatic cholangiocarcinoma (ICC) patient-derived xenograft (PDX) models was also tested in multicycle treatment and was compared to standard chemotherapy for this type of cancer (gemcitabine). The HSV-1-immunized mice significantly inhibited tumor growth in VG161-treated mice compared to control naïve treated mice. RNA expression profiling and ELISPOT analyses indicated changes in the tumor's immune profile in the immunized and treated group compared to naïve and treated mice, as well as enhanced T cell function depicted by higher numbers of tumor specific lymphocytes, which was enhanced by immunization. In the ICC PDX model, repeated treatment of VG161 with 2 or 3 cycles seemed to increase the anticancer efficacy of VG161. In conclusion, the anticancer efficacy of VG161 can be enhanced by pre-immunization with HSV-1 and multicycle administration when the virus is given intratumorally, indicating that pre-existing antiviral immunity might enhance OV-induced antitumor immunity. Our results suggest potential clinical benefits of HSV-1-based OV therapy in HSV-1-seropositive patients and multicycle administration of VG161 for long-term maintenance treatment.


Assuntos
Herpesvirus Humano 1 , Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Camundongos , Animais , Vírus Oncolíticos/fisiologia , Herpesvirus Humano 1/genética , Neoplasias/terapia , Imunidade , Modelos Animais de Doenças , Terapia Viral Oncolítica/métodos
20.
Breast Cancer Res Treat ; 125(1): 89-97, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20232140

RESUMO

Targeting HER-2 over-expressing breast cancer cells with trastuzumab has resulted in significant improvements in both disease-free and overall survival rates. However, despite a favorable initial response, some cancer cells become resistant and develop into fatal metastatic disease. Here we report that we can specifically target HER-2 over-expressing and trastuzumab-resistant breast cancer cells by using an engineered lentivirus which has trastuzumab bound to its envelope. In vitro, this lentiviral construct mediated both the expression of reporter genes, such as enhanced green fluorescent protein (EGFP) and firefly luciferase, as well as the therapeutic gene, herpes thymidine kinase (hTK), in HER-2 over-expressing cells. Subsequent application of the pro-drug ganciclovir selectively killed breast cancer cells in which lentivirus mediated expression of hTK. In vivo, we successfully targeted the expression of firefly luciferase to trastuzumab-resistant breast cancer tumors established in nude mice. Furthermore, we found that systemic administration of trastuzumab-bound lentivirus led to expression of EGFP in circulating trastuzumab-resistant breast cancer cells. In conclusion, HER-2 over-expressing breast cancer cells resistant to trastuzumab can be targeted for selective gene expression and destruction by viruses with envelope-proteins engineered to bind to this antibody.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/terapia , Resistencia a Medicamentos Antineoplásicos , Terapia Genética/métodos , Lentivirus/metabolismo , Receptor ErbB-2/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Feminino , Ganciclovir/metabolismo , Ganciclovir/farmacologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/genética , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Nus , Células Neoplásicas Circulantes/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/imunologia , Timidina Quinase/genética , Timidina Quinase/metabolismo , Trastuzumab
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA