Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163437

RESUMO

Insulin-like growth factor 1 (IGF-1) not only regulates neuronal function and development but also is neuroprotective in the setting of acute ischemic stroke. G-protein-coupled receptor 17 (GPR17) expression in brain tissue serves as an indicator of brain damage. As whether IGF-1 regulates GPR17 expression remains unknown, the aim of this study is to investigate how IGF-1 regulates GPR17 expression in vitro. Human neuroblastoma SK-N-SH cells were used. Lentivirus-mediated short hairpin RNA (shRNA) was constructed to mediate the silencing of FoxO1, while adenoviral vectors were used for its overexpression. Verification of the relevant signaling cascade was performed using a FoxO1 inhibitor (AS1842856), a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002), and a GPR17 antagonist (cangrelor). Cell proliferation was analyzed using EdU staining; immunofluorescence staining was used to detect the expression and subcellular localization of FoxO1. Chromatin immunoprecipitation was used to analyze the binding of FoxO1 to the GPR17 promoter in SK-N-SH cells. The expression of FoxO1, GPR17, and protein kinase B (also known as Akt) mRNA and protein as well as the levels of FoxO1 and Akt phosphorylation were investigated in this study. IGF-1 was found to downregulate FoxO1 and GPR17 expression in SK-N-SH cells while promoting cell viability and proliferation. Inhibition of FoxO1 and antagonism of GPR17 were found to play a role similar to that of IGF-1. Silencing of FoxO1 by lentivirus-mediated shRNA resulted in the downregulation of FoxO1 and GPR17 expression. The overexpression of FoxO1 via adenoviral vectors resulted in the upregulation of FoxO1 and GPR17 expression. Blocking of PI3K signaling by LY294002 inhibited the effect of IGF-1 on GPR17 suppression. Results from chromatin immunoprecipitation revealed that IGF-1 promotes FoxO1 nuclear export and reduces FoxO1 binding to the GPR17 promoter in SK-N-SH cells. Here, we conclude that IGF-1 enhances cell viability and proliferation in SK-N-SH cells via the promotion of FoxO1 nuclear export and reduction of FoxO1 binding to the GPR17 promoter via PI3K/Akt signaling. Our findings suggest that the enhancement of IGF-1 signaling to antagonize GPR17 serves as a potential therapeutic strategy in the management of acute ischemic stroke.


Assuntos
Regulação para Baixo , Proteína Forkhead Box O1/genética , Fator de Crescimento Insulin-Like I/metabolismo , Neurônios/citologia , Receptores Acoplados a Proteínas G/genética , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Técnicas de Inativação de Genes , Humanos , Lentivirus/fisiologia , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolonas/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Int J Mol Sci ; 22(24)2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34947988

RESUMO

Recently, we have shown the molecular basis for lactate sensing by cervical epithelial cells resulting in enhanced DNA repair processes through DNA-PKcs regulation. Interestingly, DNA-PKcs is indispensable for proper retroviral DNA integration in the cell host genome. According to recent findings, the mucosal epithelium can be efficiently transduced by retroviruses and play a pivotal role in regulating viral release by cervical epithelial cells. This study examined the effects of lactate on lentiviral transduction in cervical cancer cells (HeLa, CaSki, and C33A) and model glioma cell lines (DNA-PKcs proficient and deficient). Our study showed that L- and D-lactate enhanced DNA-PKcs presence in nuclear compartments by between 38 and 63%, which corresponded with decreased lentiviral transduction rates by between 15 and 36%. Changes in DNA-PKcs expression or its inhibition with NU7441 also greatly affected lentiviral transduction efficacy. The stimulation of cells with either HCA1 agonist 3,5-DHBA or HDAC inhibitor sodium butyrate mimicked, in part, the effects of L-lactate. The inhibition of lactate flux by BAY-8002 enhanced DNA-PKcs nuclear localization which translated into diminished lentiviral transduction efficacy. Our study suggests that L- and D-lactate present in the uterine cervix may play a role in the mitigation of viral integration in cervical epithelium and, thus, restrict the viral oncogenic and/or cytopathic potential.


Assuntos
Proteína Quinase Ativada por DNA/metabolismo , Glioma/virologia , Ácido Láctico/farmacologia , Lentivirus/fisiologia , Neoplasias do Colo do Útero/virologia , Benzoatos/farmacologia , Ácido Butírico/farmacologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Cromonas/farmacologia , Feminino , Glioma/metabolismo , Células HeLa , Humanos , Lentivirus/efeitos dos fármacos , Morfolinas/farmacologia , Transdução Genética , Neoplasias do Colo do Útero/metabolismo
3.
Virology ; 561: 6-16, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34089997

RESUMO

Based on the previously reported 13-residue antibacterial peptide analog, brevinin-1EMa (FLGWLFKVASKVL, peptide B), we attempted to design a novel class of antiviral peptides. For this goal, we synthesized three peptides with different stapling positions (B-2S, B-8S, and B-5S). The most active antiviral peptide with the specific stapling position (B-5S) was further modified in combination with either cysteine (B-5S3C, B-5S7C, and B-5S10C) or hydrophilic amino acid substitution (Bsub and Bsub-5S). Overall, B, B-5S, and Bsub-5S peptides showed superior antiviral activities against enveloped viruses such as retrovirus, lentivirus, hepatitis C virus, and herpes simplex virus with EC50 values of 1-5 µM. Murine norovirus, a non-enveloped virus, was not susceptible to the virucidal actions of these peptides, suggesting the virus membrane disruption as their main antiviral mechanisms of action. We believe that these three novel peptides could serve as promising candidates for further development of membrane-targeting antiviral drugs in the future.


Assuntos
Antivirais/farmacologia , Canais Iônicos/química , Canais Iônicos/farmacologia , Peptídeos/farmacologia , Internalização do Vírus/efeitos dos fármacos , Vírus/efeitos dos fármacos , Antivirais/química , Antivirais/metabolismo , Bactérias/efeitos dos fármacos , Linhagem Celular , Desenho de Fármacos , Hepacivirus/efeitos dos fármacos , Hepacivirus/fisiologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/fisiologia , Humanos , Canais Iônicos/metabolismo , Lentivirus/efeitos dos fármacos , Lentivirus/fisiologia , Testes de Sensibilidade Microbiana , Norovirus/efeitos dos fármacos , Norovirus/fisiologia , Peptídeos/química , Peptídeos/metabolismo , Retroviridae/efeitos dos fármacos , Retroviridae/fisiologia , Fenômenos Fisiológicos Virais
4.
J Biomed Sci ; 28(1): 34, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33926459

RESUMO

BACKGROUND: The spread of SARS-CoV-2, the virus that causes Coronavirus Disease 2019 (COVID-19), has been characterized as a worldwide pandemic. Currently, there are few preclinical animal models that suitably represent infection, as the main point of entry to human cells is via human angiotensin-converting enzyme 2 (ACE2) which is not present in typical preclinical mouse strains. Additionally, SARS-CoV-2 is highly virulent and unsafe for use in many research facilities. Here we describe the development of a preclinical animal model using intranasal administration of ACE2 followed by non-infectious SARS-CoV-2 pseudovirus (PsV) challenge. METHODS: To specifically generate our SARS-CoV-2 PsV, we used a lentivirus system. Following co-transfection with a packaging plasmid containing HIV Gag and Pol, luciferase-expressing lentiviruses, and a plasmid carrying the SARS-CoV-2 spike protein, SARS-CoV-2 PsVs can be isolated and purified. To better understand and maximize the infectivity of SARS-CoV-2 PsV, we generated PsV carrying spike protein variants known to have varying human ACE2 binding properties, including 19 deletion (19del) and 19del + D614G. RESULTS: Our system demonstrated the ability of PsVs to infect the respiratory passage of mice following intranasal hACE2 transduction. Additionally, we demonstrate in vitro and in vivo manipulability of our system using recombinant receptor-binding domain protein to prevent PsV infection. CONCLUSIONS: Our PsV system is able to model SARS-CoV-2 infections in a preclinical mouse model and can be used to test interventions or preventative treatments. We believe that this method can be extended to work in various mouse strains or to model infection with different coronaviruses. A simple in vivo system such as our model is crucial for rapidly and effectively responding to the current COVID-19 pandemic in addition to preparing for future potential coronavirus outbreaks.


Assuntos
Enzima de Conversão de Angiotensina 2/administração & dosagem , COVID-19 , Modelos Animais de Doenças , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/fisiologia , Administração Intranasal , Animais , COVID-19/prevenção & controle , COVID-19/transmissão , COVID-19/virologia , Feminino , Humanos , Lentivirus/fisiologia , Camundongos , Camundongos Endogâmicos BALB C
5.
Viruses ; 13(1)2021 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-33477360

RESUMO

The APOBEC3 family of proteins in mammals consists of cellular cytosine deaminases and well-known restriction factors against retroviruses, including lentiviruses. APOBEC3 genes are highly amplified and diversified in mammals, suggesting that their evolution and diversification have been driven by conflicts with ancient viruses. At present, lentiviruses, including HIV, the causative agent of AIDS, are known to encode a viral protein called Vif to overcome the antiviral effects of the APOBEC3 proteins of their hosts. Recent studies have revealed that the acquisition of an anti-APOBEC3 ability by lentiviruses is a key step in achieving successful cross-species transmission. Here, we summarize the current knowledge of the interplay between mammalian APOBEC3 proteins and viral infections and introduce a scenario of the coevolution of mammalian APOBEC3 genes and viruses.


Assuntos
Desaminases APOBEC/metabolismo , Interações Hospedeiro-Patógeno , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Retroviridae/fisiologia , Desaminases APOBEC/genética , Animais , Resistência à Doença/genética , Evolução Molecular , Variação Genética , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Humanos , Lentivirus/fisiologia , Filogenia , Infecções por Retroviridae/transmissão , Especificidade da Espécie , Produtos do Gene vif do Vírus da Imunodeficiência Humana
6.
Nat Biomed Eng ; 5(2): 144-156, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33398131

RESUMO

Therapeutic genome editing requires effective and targeted delivery methods. The delivery of Cas9 mRNA using adeno-associated viruses has led to potent in vivo therapeutic efficacy, but can cause sustained Cas9 expression, anti-Cas9 immune responses and off-target edits. Lentiviral vectors have been engineered to deliver nucleases that are expressed transiently, but in vivo evidence of their biomedical efficacy is lacking. Here, we show that the lentiviral codelivery of Streptococcus pyogenes Cas9 mRNA and expression cassettes that encode a guide RNA that targets vascular endothelial growth factor A (Vegfa) is efficacious in a mouse model of wet age-related macular degeneration induced by Vegfa. A single subretinal injection of engineered lentiviruses knocked out 44% of Vegfa in retinal pigment epithelium and reduced the area of choroidal neovascularization by 63% without inducing off-target edits or anti-Cas9 immune responses. Engineered lentiviruses for the transient expression of nucleases may form the basis of new treatments for retinal neovascular diseases.


Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Edição de Genes/métodos , Lentivirus/fisiologia , Degeneração Macular/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Modelos Animais de Doenças , Vetores Genéticos/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética
7.
Methods Mol Biol ; 2254: 323-338, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33326085

RESUMO

CRISPR-mediated interference (CRISPRi), a robust and specific system for programmably repressing transcription, provides a versatile tool for systematically characterizing the function of long noncoding RNAs (lncRNAs). When used with highly parallel, lentiviral pooled screening approaches, CRISPRi enables the targeted knockdown of tens of thousands of lncRNA-expressing loci in a single screen. Here we describe the use of CRISPRi to target lncRNA loci in a pooled screen, using cell growth and proliferation as an example of a phenotypic readout. Considerations for custom lncRNA-targeting libraries, alternative phenotypic readouts, and orthogonal validation approaches are also discussed.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Lentivirus/fisiologia , RNA Longo não Codificante/genética , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Células HEK293 , Humanos , Lentivirus/genética , Regiões Promotoras Genéticas , Transcrição Gênica
8.
STAR Protoc ; 1(3): 100174, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33377068

RESUMO

Glioblastoma (GBM) is the most common malignant adult brain tumor that is resistant to the standard care therapy. Advances in chimeric antigen receptor (CAR) T cell therapies have spurred renewed interest in developing CAR T cell therapies to target chemoradiotherapy-resistant brain tumor-initiating cells. This protocol shows how to isolate peripheral blood mononuclear cells from healthy donors and generate CAR T cells for the antigens of interest, and how to intracranially inject the CAR T cells into a patient-derived xenograft mouse model of GBM. For complete details on the use and execution of this protocol, please refer to Vora et al. (2020).


Assuntos
Glioblastoma/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Pontos de Referência Anatômicos , Animais , Proliferação de Células , Células HEK293 , Humanos , Lentivirus/fisiologia , Ativação Linfocitária/imunologia , Camundongos , Células-Tronco Neoplásicas/patologia , Plasmídeos/metabolismo
9.
Reprod Sci ; 27(11): 2082-2091, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32542539

RESUMO

Endometrial-like stromal cells, one of the main components of endometriotic lesions, are an important in vitro model for studying cellular and molecular mechanisms associated with lesion development in endometriosis. However, the short life span of primary endometriotic stromal cells (Ec-ESCs) limits their use. Human telomerase reverse transcriptase (hTERT) plasmids can be used to develop immortalized cell lines. Here we aimed to establish an endometriotic stromal cell line by hTERT immortalization. Primary Ec-ESCs were obtained from a human ovarian endometriotic cyst. The purity was assessed by morphology and the expression of vimentin, cytokeratin, and human interferon-inducible transmembrane protein 1 (hIFITM1). Cells were infected with hTERT lentiviral vector and selected with hygromycin. hTERT mRNA levels were confirmed by RT-qPCR. Immortalized Ec-ESCs (iEc-ESCs) were characterized by examining the expression of morphological markers and key genes of interest, TP53, estrogen receptor ß (ERß), progesterone receptor (PR), and steroidogenic factor-1 (SF-1). Karyotyping and in vitro decidualization studies were also performed. Ec-ESCs were positive for vimentin and hIFITM1 and negative for cytokeratin, indicating that they were representative of Ec-ESC. The fibroblast-like morphology, expression of TP53, ERß, PR, and SF-1 did not change before and after hTERT immortalization. iEc-ESCs showed an impaired decidualization response like primary Ec-ESCs when compared to normal eutopic stromal cells. Karyotyping showed that 15/19 cells had normal female karyotype, while 4/19 cells had partial trisomy 11q. Collectively, we successfully established and characterized an immortalized endometriotic stromal cell line. It is potentially useful as an in vitro experimental model to investigate endometriosis biology.


Assuntos
Técnicas de Cultura de Células/métodos , Endometriose/fisiopatologia , Endométrio/fisiologia , Células Estromais/fisiologia , Linhagem Celular , Feminino , Vetores Genéticos , Humanos , Lentivirus/fisiologia , Plasmídeos , Telomerase
10.
Virol J ; 17(1): 22, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32039735

RESUMO

Transfection, the process of introducing purified nucleic acids into cells, and viral transduction, viral-mediated nucleic acid transfer, are two commonly utilized techniques for gene delivery in the research setting. Transfection allows purified nucleic acid to be introduced into target cells through chemical-based techniques, nonchemical methods or particle-based methods, while viral transduction employs genomes or vectors based on adenoviruses, retroviruses (e.g. lentiviruses), adeno-associated viruses, or hybrid viruses. Transfected DNAs are often tested for potential effects on subsequent transduction, but it is not clear whether transfection itself rather than the particular nucleic acid being introduced might impact subsequent viral transfection. We observed a significant association between successfully transfected mobilized peripheral blood CD34+ human stem and progenitor cells (HSPCs) and permissiveness to subsequent lentiviral transduction, which was not evident in other cells such as 293 T cells and Jurkat cells. This association, apparently specific to CD34+ human stem and progenitor cells (HSPCs), is critical to both research and clinical applications as these cells are a frequent target of transfection and viral transduction owing to the durable nature of these cells in living systems. This finding may also present a significant opportunity to enhance the success of viral transduction for clinical applications.


Assuntos
Células-Tronco Hematopoéticas/virologia , Lentivirus/genética , Lentivirus/fisiologia , Transdução Genética , Transfecção , Antígenos CD34 , Vetores Genéticos , Células HEK293 , Humanos , Células Jurkat
11.
Methods Mol Biol ; 2150: 121-129, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31020637

RESUMO

Stem cell-based therapies hold great promise as alternative therapeutic strategies for various chronic diseases, including ischemic cardiomyopathy. Tracking the engraftment of transplanted stem cells is critical to the assessment of donor cell survival in the host environment. Fluorescent proteins, such as green fluorescent protein (GFP), have been widely used to track the fate of donor cells; however, GFP labeling has limitations with regard to noninvasively measuring cell engraftment in vivo. Our research indicates that near-infrared fluorescent protein (iRFP) labeling offers advantages for noninvasive in vivo imaging and histological assessment. Here, we present a protocol for using the lentiviral vector-mediated iRFP vector system to label and track donor stem cells in ischemic mouse hearts.


Assuntos
Rastreamento de Células/métodos , Imageamento Tridimensional , Raios Infravermelhos , Proteínas Luminescentes/metabolismo , Isquemia Miocárdica/diagnóstico por imagem , Coloração e Rotulagem , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/metabolismo , Lentivirus/fisiologia , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Empacotamento do Genoma Viral
12.
J Virol Methods ; 276: 113768, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31704112

RESUMO

Lentiviral vectors and lentiviruses are important tools for basic and applied biomedical research. Yet, biosafety regulations from legal authorities have to be fulfilled when transferring BSL-2 to -3 vectors/viruses to facilities with lower biosafety level. Here, we (re-)evaluated different chemical and thermal approaches to inactivate vesicular stomatitis virus G-protein (VSV-G) pseudotyped lentiviral vectors and either wildtype or VSV-G pseudotyped human immunodeficiency viruses (HIV). Aldehydes, detergents and alcohols were as effective as thermal inactivation procedures to efficiently inactivate purified lentiviral vectors and replication-competent HIV. In addition, no residual infectivity was detected when inactivating HIV-infected TZM-bl reporter cells with selected detergents and aldehydes. Thus, our established inactivation protocols can be used by other laboratories working with lentiviral vectors or infectious lentiviruses and provide a template for viruses with similar physicochemical properties.


Assuntos
Vetores Genéticos/efeitos dos fármacos , HIV/efeitos dos fármacos , Lentivirus/efeitos dos fármacos , Inativação de Vírus/efeitos dos fármacos , Álcoois/farmacologia , Aldeídos/farmacologia , Detergentes/farmacologia , Células HEK293 , HIV/patogenicidade , Temperatura Alta , Humanos , Lentivirus/fisiologia
13.
Sci Rep ; 9(1): 15745, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31673064

RESUMO

The objective of this study was to establish a versatile cell line for replication-incompetent virus production and inactivation with formaldehyde to generate a model of cell-based vaccine manufacturing process. To achieve this goal, we took advantage of the easily accessed chick embryonic fibroblasts. Nine-day old chick embryonic fibroblasts were obtained and subjected to be transduced with a set of lentivirus to develop a chick induced pluripotent stem (ciPS) cell line. Morphological features, positive periodic acid-Schiff staining as well as strong immunocytofluorescence of alkaline phosphatase, intestinal (ALPI) and POU class 5 homeobox 1 (POU5F1) proteins suggested that these chick embryonic fibroblasts have been transformed into ciPS cells. Further differentiation and immunocytofluorescence assays confirmed that this ciPS cell line possesses capacities and potentials to form embryoid bodies, differentiate into all three embryonic layers: ectoderm, mesoderm and endoderm with evidence of strongly positive and specific molecular markers. Immunoblot analysis next demonstrated that through recombinant DNA technology and the 2nd generation lentiviral transfer system, the goose hemagglutinin gene (H5) gene was packaged into the replication-incompetent virus and highly expressed in a bladder cancer-derived cell line, T24, after transduction. The titer of ciPS-generated replication-incompetent virus is comparable to that from the Phoenix-AMPHO cell line, which is a commercial and high productive retrovirus producer. Our study successfully established a ciPS cell line which is able to produce replication-incompetent virus, providing a new strategy for cell-based vaccine production after virus inactivation.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Lentivirus/genética , Plasmídeos/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Embrião de Galinha , Galinhas , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Gansos , Hemaglutininas/genética , Hemaglutininas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Lentivirus/fisiologia , Fator 3 de Transcrição de Octâmero/metabolismo , Plasmídeos/genética , Taiwan , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/isolamento & purificação , Vacinas Sintéticas/metabolismo , Replicação Viral
14.
J Virol ; 93(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31167917

RESUMO

Lentiviral replication mediated by reverse transcriptase is considered to be highly error prone, leading to a high intra-individual evolution rate that promotes evasion of neutralization and persistent infection. Understanding lentiviral intra-individual evolutionary dynamics on a comparative basis can therefore inform research strategies to aid in studies of pathogenesis, vaccine design, and therapeutic intervention. We conducted a systematic review of intra-individual evolution rates for three species groups of lentiviruses-feline immunodeficiency virus (FIV), simian immunodeficiency virus (SIV), and human immunodeficiency virus (HIV). Overall, intra-individual rate estimates differed by virus but not by host, gene, or viral strain. Lentiviral infections in spillover (nonadapted) hosts approximated infections in primary (adapted) hosts. Our review consistently documents that FIV evolution rates within individuals are significantly lower than the rates recorded for HIV and SIV. FIV intra-individual evolution rates were noted to be equivalent to FIV interindividual rates. These findings document inherent differences in the evolution of FIV relative to that of primate lentiviruses, which may signal intrinsic difference of reverse transcriptase between these viral species or different host-viral interactions. Analysis of lentiviral evolutionary selection pressures at the individual versus population level is valuable for understanding transmission dynamics and the emergence of virulent and avirulent strains and provides novel insight for approaches to interrupt lentiviral infections.IMPORTANCE To the best of our knowledge, this is the first study that compares intra-individual evolution rates for FIV, SIV, and HIV following systematic review of the literature. Our findings have important implications for informing research strategies in the field of intra-individual virus dynamics for lentiviruses. We observed that FIV evolves more slowly than HIV and SIV at the intra-individual level and found that mutation rates may differ by gene sequence length but not by host, gene, strain, an experimental setting relative to a natural setting, or spillover host infection relative to primary host infection.


Assuntos
Evolução Biológica , Interações Hospedeiro-Patógeno , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Animais , Gatos , Evolução Molecular , Síndrome de Imunodeficiência Adquirida Felina/virologia , Variação Genética , HIV/genética , Infecções por HIV/virologia , Humanos , Vírus da Imunodeficiência Felina/genética , Lentivirus/classificação , Primatas , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética
15.
Oncol Rep ; 42(1): 370-376, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115558

RESUMO

The present study aimed to investigate the antitumor effects of an intratumoral injection of dendritic cells (DCs) overexpressing interleukin-12 (IL­12) on melanoma. DCs, isolated from mouse spleen, were gene­modified using an IL­12 overexpression vector. Melanoma B6 cells were injected into C57BL/6 mice to generate tumors. Thereafter, DCs overexpressing IL­12 were injected into the tumors, and tumor volume was subsequently measured. Pathological changes in tumor tissue were detected by hematoxylin and eosin staining. The expression of interleukin-4 (IL­4) and IL­12 in tumors was measured by enzyme­linked immunosorbent assay, real­time PCR and western blotting. DCs were successfully isolated and a lentivirus vector expressing IL­12 was constructed. After intratumoral injection of phosphate­buffered saline (control group), tumor cells exhibited malignant growth; whereas tumors injected with DCs (DC group) or DCs + empty vector (DC + vector group) exhibited a small amount of inflammatory cell infiltration and limited areas of tissue necrosis. In contrast, tumors injected with DCs overexpressing IL­12 (DC + IL­12 group) displayed severe tissue necrosis, loss of cell structure, and inflammatory cell infiltration. Compared with the control group, the tumor volumes were significantly lower in the DC, the DC + vector and the DC + IL­12 groups, while the expression of IL­12 and IL­4 in the tumors was significantly higher. Importantly, the most marked changes in tumor volume and IL­12 and IL­4 expression were in the DC + IL­12 group, which were significantly greater than those in tumors treated with unmodified DCs. Hence, intratumoral injection of DCs overexpressing IL­12 exerted strong antitumor effects in melanoma, and biotherapy with DCs overexpressing IL­12 is a potential treatment strategy for melanoma.


Assuntos
Células Dendríticas/transplante , Interleucina-12/metabolismo , Lentivirus/fisiologia , Melanoma Experimental/terapia , Animais , Linhagem Celular Tumoral , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Injeções Intralesionais , Interleucina-12/genética , Interleucina-4/genética , Interleucina-4/metabolismo , Lentivirus/genética , Masculino , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Transfecção , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS Comput Biol ; 15(3): e1006773, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30835721

RESUMO

Tumor therapy with replication competent viruses is an exciting approach to cancer eradication where viruses are engineered to specifically infect, replicate, spread and kill tumor cells. The outcome of tumor virotherapy is complex due to the variable interactions between the cancer cell and virus populations as well as the immune response. Oncolytic viruses are highly efficient in killing tumor cells in vitro, especially in a 2D monolayer of tumor cells, their efficiency is significantly lower in a 3D environment, both in vitro and in vivo. This indicates that the spatial dimension may have a major influence on the dynamics of virus spread. We study the dynamic behavior of a spatially explicit computational model of tumor and virus interactions using a combination of in vitro 2D and 3D experimental studies to inform the models. We determine the number of nearest neighbor tumor cells in 2D (median = 6) and 3D tumor spheroids (median = 16) and how this influences virus spread and the outcome of therapy. The parameter range leading to tumor eradication is small and even harder to achieve in 3D. The lower efficiency in 3D exists despite the presence of many more adjacent cells in the 3D environment that results in a shorter time to reach equilibrium. The mean field mathematical models generally used to describe tumor virotherapy appear to provide an overoptimistic view of the outcomes of therapy. Three dimensional space provides a significant barrier to efficient and complete virus spread within tumors and needs to be explicitly taken into account for virus optimization to achieve the desired outcome of therapy.


Assuntos
Simulação por Computador , Modelos Biológicos , Neoplasias/terapia , Terapia Viral Oncolítica , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Lentivirus/fisiologia , Vírus do Sarampo/fisiologia , Neoplasias/patologia , Esferoides Celulares/patologia , Microambiente Tumoral , Replicação Viral
17.
Methods Mol Biol ; 1937: 125-134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30706393

RESUMO

Lentiviral vectors (LVs) are widely used in gene transfer protocols due to many advantages that include stable gene expression, higher transgene payloads, and, importantly, the ability to pseudotype the vectors with a diverse number of heterologous viral envelopes with broad or restricted cell tropism depending on the need. The pseudotyping process also allows for incorporation of specific antibodies/ligands to engineer LVs. These features greatly facilitate customization of lentiviral vectors for cell/tissue specific gene delivery. The VSV-G protein containing envelope remains the most widely used among the viral glycoproteins used for LV pseudotyping due to its versatile host range and stability. However, many other viral envelopes are being identified for special applications of LVs. Here we describe the methodology to generate pseudotyped LVs using a four-plasmid transient transfection system focusing on aspects to generate high-titer vector stocks.


Assuntos
Lentivirus/fisiologia , Plasmídeos/genética , Proteínas do Envelope Viral/metabolismo , Cultura de Vírus/métodos , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas do Envelope Viral/genética , Carga Viral , Tropismo Viral , Montagem de Vírus
18.
Biotechnol Genet Eng Rev ; 35(1): 26-45, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30317930

RESUMO

Alzheimer's disease (AD) is the most common form of dementia and has affected millions of individuals worldwide. The hallmarks of AD include the amyloid beta plaque deposits, tau neurofibrillary tangles, altered neuronal signaling, alongside decline in memory and cognitive functions. Conventional drug therapies do exist, such as donepezil or aducanumab, but these drugs mostly focus on halting AD progression instead of causing a reversal within the disease. In an effort to ameliorate and ultimately cure AD, researchers have delved into viral-mediated gene therapy to fix this disease from its root molecular causes. To date, adeno-associated virus and lentiviral vectors have remained the most vastly studied among other viral vectors to combat AD. These vectors could be employed alongside various genetic materials based on the types of processes we want to alter to yield a positive effect, such as disruption of amyloidogenic pathway, neuroprotection and lipid metabolism pathways. Recent studies and trials were reviewed in this article, highlighting their clinical significance, differences and limitations between each method. By learning from the different combinations and possibilities of viral-mediated gene transfer, researchers would then get a step closer in ameliorating symptoms and possibly in curing AD.


Assuntos
Doença de Alzheimer/terapia , Dependovirus/fisiologia , Terapia Genética/métodos , Lentivirus/fisiologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Ensaios Clínicos como Assunto , Dependovirus/genética , Progressão da Doença , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Lentivirus/genética , Redes e Vias Metabólicas , Resultado do Tratamento
19.
Nat Commun ; 9(1): 5193, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518925

RESUMO

Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/imunologia , Infecções por Lentivirus/imunologia , Monócitos/imunologia , NF-kappa B/imunologia , Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Células Cultivadas , Humanos , Terapia de Imunossupressão , Lentivirus/fisiologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/fisiopatologia , Infecções por Lentivirus/virologia , Células Mieloides/imunologia , NF-kappa B/genética
20.
Viruses ; 10(11)2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30405009

RESUMO

Recent developments in defining the role of the lentiviral envelope glycoprotein (Env) cytoplasmic tail (CT) in Env trafficking and incorporation into virus particles have advanced our understanding of viral replication and transmission. To stimulate additional progress in this field, the two-day International Workshop on Structure and Function of the Lentiviral gp41 Cytoplasmic Tail, co-organized by Eric Freed and James Hoxie, was held at the National Cancer Institute in Frederick, MD (26⁻27 April 2018). The meeting served to bring together experts focused on the role of gp41 in HIV replication and to discuss the emerging mechanisms of CT-dependent trafficking, Env conformation and structure, host protein interaction, incorporation, and viral transmission. The conference was organized around the following three main hot topics in gp41 research: the role of host factors in CT-dependent Env incorporation, Env structure, and CT-mediated trafficking and transmission. This review highlights important topics and the advances in gp41 research that were discussed during the conference.


Assuntos
Proteína gp41 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/metabolismo , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Animais , Proteína gp41 do Envelope de HIV/genética , Interações Hospedeiro-Patógeno , Humanos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA