Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell ; 134(4): 577-86, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18691745

RESUMO

Evaluation of the therapeutic potential of RNAi for HIV infection has been hampered by the challenges of siRNA delivery and lack of suitable animal models. Using a delivery method for T cells, we show that siRNA treatment can dramatically suppress HIV infection. A CD7-specific single-chain antibody was conjugated to oligo-9-arginine peptide (scFvCD7-9R) for T cell-specific siRNA delivery in NOD/SCIDIL2rgamma-/- mice reconstituted with human lymphocytes (Hu-PBL) or CD34+ hematopoietic stem cells (Hu-HSC). In HIV-infected Hu-PBL mice, treatment with anti-CCR5 (viral coreceptor) and antiviral siRNAs complexed to scFvCD7-9R controlled viral replication and prevented the disease-associated CD4 T cell loss. This treatment also suppressed endogenous virus and restored CD4 T cell counts in mice reconstituted with HIV+ peripheral blood mononuclear cells. Moreover, scFvCD7-9R could deliver antiviral siRNAs to naive T cells in Hu-HSC mice and effectively suppress viremia in infected mice. Thus, siRNA therapy for HIV infection appears to be feasible in a preclinical animal model.


Assuntos
Infecções por HIV/genética , Infecções por HIV/terapia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Linfócitos T/metabolismo , Animais , Antígenos CD7/metabolismo , Modelos Animais de Doenças , Expressão Gênica , HIV-1/genética , HIV-1/metabolismo , Humanos , Fragmentos de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , RNA Viral/metabolismo
3.
Eur J Immunol ; 45(1): 82-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25270431

RESUMO

Targeting DNA vaccines to dendritic cells (DCs) greatly enhances immunity. Although several approaches have been used to target protein Ags to DCs, currently there is no method that targets DNA vaccines directly to DCs. Here, we show that a small peptide derived from the rabies virus glycoprotein fused to protamine residues (RVG-P) can target DNA to myeloid cells, including DCs, which results in enhanced humoral and T-cell responses. DCs targeted with a DNA vaccine encoding the immunodominant vaccinia B8R gene via RVG-P were able to restimulate vaccinia-specific memory T cells in vitro. Importantly, a single i.v. injection of B8R gene bound to RVG-P was able to prime a vaccinia-specific T-cell response that was able to rapidly clear a subsequent vaccinia challenge in mice. Moreover, delivery of DNA in DCs was enough to induce DC maturation and efficient Ag presentation without the need for adjuvants. Finally, immunization of mice with a DNA-vaccine encoding West Nile virus (WNV) prM and E proteins via RVG-P elicited high titers of WNV-neutralizing Abs that protected mice from lethal WNV challenge. Thus, RVG-P provides a reagent to target DNA vaccines to myeloid cells and elicit robust T-cell and humoral immune responses.


Assuntos
Células Dendríticas/imunologia , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Vacinas de DNA/administração & dosagem , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/efeitos dos fármacos , Antígenos Virais/genética , Antígenos Virais/imunologia , Células Dendríticas/citologia , Células Dendríticas/virologia , Engenharia Genética , Glicoproteínas/genética , Glicoproteínas/imunologia , Imunização , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peptídeos/administração & dosagem , Peptídeos/síntese química , Vírus da Raiva/genética , Vírus da Raiva/imunologia , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/virologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia
4.
Mol Ther ; 23(10): 1663-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26061648

RESUMO

Attempts at eliciting neutralizing antibodies against human immunodeficiency virus (HIV)-1 have generally failed. Computationally designed epitope-scaffold platforms allow transplantation of structural epitopes to scaffold proteins. Human rhinovirus (HRV) allows such engrafting of HIV-1 epitopes on the surface scaffold proteins. However, since HRV infects only humans and great apes, the efficacy of chimeric HRV-based live viral vaccines is difficult to assess in animal models. Here, we used human ICAM-1 transgenic (hICAM-1 Tg) mice that support productive HRV infection to assess the efficacy of chimeric HRV expressing the HIV-1 membrane proximal external region (MPER) epitope, 4E10. Intranasal immunization with chimeric HRV in transgenic mice effectively induced antibodies that recognized 4E10 peptide as well as HIV-1 Env trimer. Importantly, the immunized mouse sera were able to neutralize HIV strains including those belonging to clades B and C. Moreover, intranasal immunization could bypass pre-existing immunity to HRV. Thus, chimeric HRV appears to provide a viable vaccine vehicle for HIV-1 immunization in humans.


Assuntos
Anticorpos Neutralizantes/imunologia , Epitopos/imunologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/imunologia , Rhinovirus/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/química , Apresentação de Antígeno/imunologia , Modelos Animais de Doenças , Epitopos/química , Epitopos/genética , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/química , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/genética , Humanos , Imunização , Molécula 1 de Adesão Intercelular/genética , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Ligação Proteica/imunologia , Conformação Proteica
5.
Mol Ther ; 23(2): 310-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25358251

RESUMO

Multiplexed miRNA-based shRNAs (shRNA-miRs) could have wide potential to simultaneously suppress multiple genes. Here, we describe a simple strategy to express a large number of shRNA-miRs using minimal flanking sequences from multiple endogenous miRNAs. We found that a sequence of 30 nucleotides flanking the miRNA duplex was sufficient for efficient processing of shRNA-miRs. We inserted multiple shRNAs in tandem, each containing minimal flanking sequence from a different miRNA. Deep sequencing of transfected cells showed accurate processing of individual shRNA-miRs and that their expression did not decrease with the distance from the promoter. Moreover, each shRNA was as functionally competent as its singly expressed counterpart. We used this system to express one shRNA-miR targeting CCR5 and six shRNA-miRs targeting the HIV-1 genome. The lentiviral construct was pseudotyped with HIV-1 envelope to allow transduction of both resting and activated primary CD4 T cells. Unlike one shRNA-miR, the seven shRNA-miR transduced T cells nearly abrogated HIV-1 infection in vitro. Additionally, when PBMCs from HIV-1 seropositive individuals were transduced and transplanted into NOD/SCID/IL-2R γc(-/-) mice (Hu-PBL model) efficient suppression of endogenous HIV-1 replication with restoration of CD4 T cell counts was observed. Thus, our multiplexed shRNA appears to provide a promising gene therapeutic approach for HIV-1 infection.


Assuntos
Infecções por HIV/virologia , HIV-1/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Replicação Viral/genética , Animais , Contagem de Linfócito CD4 , Linhagem Celular , Modelos Animais de Doenças , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/genética , Infecções por HIV/imunologia , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Camundongos , Receptores CCR5/genética , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/virologia , Transdução Genética
6.
Clin Immunol ; 156(1): 58-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25463432

RESUMO

Although patients with GVHD have elevated serum levels of IL10, whether its role is protective or pathogenic remains unclear. Here, we used a humanized mouse model to study the role of IL-10 in GVHD. When human PBMCs were engrafted in NOD-scid IL2rγc(null) mice expressing human IL-10, the T cells underwent massive expansion resulting in lethality by day 21, whereas control mice survived for at least 40 days. Histopathology of the liver showed extensive mononuclear cell infiltration in IL-10 expressing but not in control mice. Corresponding to their aggressiveness, the T cells in the IL-10 group exhibited predominantly an effector memory phenotype (CD45RO(+)CD27(-)) while in control mice, the T cells were of transitional memory phenotype (CD45RO(+)CD27(+)). Further, IL-10 receptor blocking antibody was able to protect the animals from GVHD. Since our results demonstrate a direct pathogenic role for IL-10, blockade of IL-10 signaling may provide a therapeutic option for GVHD.


Assuntos
Doença Enxerto-Hospedeiro/fisiopatologia , Interleucina-10/metabolismo , Linfócitos T/citologia , Animais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Modelos Animais
7.
Proc Natl Acad Sci U S A ; 109(51): 21052-7, 2012 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-23213216

RESUMO

Hypersecretion of cytokines by innate immune cells is thought to initiate multiple organ failure in murine models of sepsis. Whether human cytokine storm also plays a similar role is not clear. Here, we show that human hematopoietic cells are required to induce sepsis-induced mortality following cecal ligation and puncture (CLP) in the severely immunodeficient nonobese diabetic (NOD)/SCID/IL2Rγ(-/-) mice, and siRNA treatment to inhibit HMGB1 release by human macrophages and dendritic cells dramatically reduces sepsis-induced mortality. Following CLP, compared with immunocompetent WT mice, NOD/SCID/IL2Rγ(-/-) mice did not show high levels of serum HMGB1 or murine proinflammatory cytokines and were relatively resistant to sepsis-induced mortality. In contrast, NOD/SCID/IL2Rγ(-/-) mice transplanted with human hematopoietic stem cells [humanized bone marrow liver thymic mice (BLT) mice] showed high serum levels of HMGB1, as well as multiple human but not murine proinflammatory cytokines, and died uniformly, suggesting human cytokines are sufficient to induce organ failure in this model. Moreover, targeted delivery of HMGB1 siRNA to human macrophages and dendritic cells using a short acetylcholine receptor (AchR)-binding peptide [rabies virus glycoprotein (RVG)-9R] effectively suppressed secretion of HMGB1, reduced the human cytokine storm, human lymphocyte apoptosis, and rescued humanized mice from CLP-induced mortality. siRNA treatment was also effective when started after the appearance of sepsis symptoms. These results show that CLP in humanized mice provides a model to study human sepsis, HMGB1 siRNA might provide a treatment strategy for human sepsis, and RVG-9R provides a tool to deliver siRNA to human macrophages and dendritic cells that could potentially be used to suppress a variety of human inflammatory diseases.


Assuntos
Células Dendríticas/citologia , Proteína HMGB1/metabolismo , Macrófagos/citologia , Sepse/metabolismo , Animais , Citocinas/metabolismo , Inativação Gênica , Técnicas de Transferência de Genes , Humanos , Sistema Imunitário , Inflamação , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Peptídeos/química , RNA Interferente Pequeno/metabolismo
8.
Nature ; 448(7149): 39-43, 2007 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-17572664

RESUMO

A major impediment in the treatment of neurological diseases is the presence of the blood-brain barrier, which precludes the entry of therapeutic molecules from blood to brain. Here we show that a short peptide derived from rabies virus glycoprotein (RVG) enables the transvascular delivery of small interfering RNA (siRNA) to the brain. This 29-amino-acid peptide specifically binds to the acetylcholine receptor expressed by neuronal cells. To enable siRNA binding, a chimaeric peptide was synthesized by adding nonamer arginine residues at the carboxy terminus of RVG. This RVG-9R peptide was able to bind and transduce siRNA to neuronal cells in vitro, resulting in efficient gene silencing. After intravenous injection into mice, RVG-9R delivered siRNA to the neuronal cells, resulting in specific gene silencing within the brain. Furthermore, intravenous treatment with RVG-9R-bound antiviral siRNA afforded robust protection against fatal viral encephalitis in mice. Repeated administration of RVG-9R-bound siRNA did not induce inflammatory cytokines or anti-peptide antibodies. Thus, RVG-9R provides a safe and noninvasive approach for the delivery of siRNA and potentially other therapeutic molecules across the blood-brain barrier.


Assuntos
Encéfalo , Sistemas de Liberação de Medicamentos , Glicoproteínas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Sequência de Aminoácidos , Animais , Barreira Hematoencefálica , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular , Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa/prevenção & controle , Inativação Gênica , Vetores Genéticos/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Lentivirus/genética , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Dados de Sequência Molecular , Neurônios/metabolismo , Neurônios/virologia , Oligopeptídeos/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Vírus da Raiva/genética , Vírus da Raiva/fisiologia , Receptores Nicotínicos/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Proteínas Virais/administração & dosagem , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
J Control Release ; 359: 26-32, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37236320

RESUMO

The CXCR4 chemokine is a key molecular regulator of many biological functions controlling leukocyte functions during inflammation and immunity, and during embryonic development. Overexpression of CXCR4 is also associated with many types of cancer where its activation promotes angiogenesis, tumor growth/survival, and metastasis. In addition, CXCR4 is involved in HIV replication, working as a co-receptor for viral entry, making CXCR4 a very attractive target for developing novel therapeutic agents. Here we report the pharmacokinetic profile in rats of a potent CXCR4 antagonist cyclotide, MCo-CVX-5c, previously developed in our group that displayed a remarkable in vivo resistance to biological degradation in serum. This bioactive cyclotide, however, was rapidly eliminated through renal clearance. Several lipidated versions of cyclotide MCo-CVX-5c showed a significant increase in the half-life when compared to the unlipidated form. The palmitoylated version of cyclotide MCo-CVX-5c displayed similar CXCR4 antagonistic activity as the unlipidated cyclotide, while the cyclotide modified with octadecanedioic (18-oxo-octadecanoic) acid exhibited a remarkable decrease in its ability to antagonize CXCR4. Similar results were also obtained when tested for its ability to inhibit growth in two cancer cell lines and HIV infection in cells. These results show that the half-life of cyclotides can be improved by lipidation although it can also affect their biological activity depending on the lipid employed.


Assuntos
Ciclotídeos , Infecções por HIV , Neoplasias , Ratos , Animais , Ciclotídeos/farmacologia , Linhagem Celular , Receptores CXCR4
10.
J Virol ; 84(5): 2490-501, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20015996

RESUMO

Dengue is a common arthropod-borne flaviviral infection in the tropics, for which there is no vaccine or specific antiviral drug. The infection is often associated with serious complications such as dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS), in which both viral and host factors have been implicated. RNA interference (RNAi) is a potent antiviral strategy and a potential therapeutic option for dengue if a feasible strategy can be developed for delivery of small interfering RNA (siRNA) to dendritic cells (DCs) and macrophages, the major in vivo targets of the virus and also the source of proinflammatory cytokines. Here we show that a dendritic cell-targeting 12-mer peptide (DC3) fused to nona-D-arginine (9dR) residues (DC3-9dR) delivers siRNA and knocks down endogenous gene expression in heterogenous DC subsets, (monocyte-derived DCs [MDDCs], CD34(+) hematopoietic stem cell [HSC])-derived Langerhans DCs, and peripheral blood DCs). Moreover, DC3-9dR-mediated delivery of siRNA targeting a highly conserved sequence in the dengue virus envelope gene (siFvE(D)) effectively suppressed dengue virus replication in MDDCs and macrophages. In addition, DC-specific delivery of siRNA targeting the acute-phase cytokine tumor necrosis factor alpha (TNF-alpha), which plays a major role in dengue pathogenesis, either alone or in combination with an antiviral siRNA, significantly reduced virus-induced production of the cytokine in MDDCs. Finally to validate the strategy in vivo, we tested the ability of the peptide to target human DCs in the NOD/SCID/IL-2Rgamma(-/-) mouse model engrafted with human CD34(+) hematopoietic stem cells (HuHSC mice). Treatment of mice by intravenous (i.v.) injection of DC3-9dR-complexed siRNA targeting TNF-alpha effectively suppressed poly(I:C)-induced TNF-alpha production by DCs. Thus, DC3-9dR can deliver siRNA to DCs both in vitro and in vivo, and this delivery approach holds promise as a therapeutic strategy to simultaneously suppress virus replication and curb virus-induced detrimental host immune responses in dengue infection.


Assuntos
Citocinas/biossíntese , Células Dendríticas , Vírus da Dengue , Dengue , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Animais , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Dengue/imunologia , Dengue/terapia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/imunologia , Técnicas de Transferência de Genes , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Oligopeptídeos/genética , Oligopeptídeos/imunologia , Peptídeos/genética , Peptídeos/metabolismo , Poli I-C/imunologia , RNA Interferente Pequeno/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Replicação Viral/efeitos dos fármacos
11.
Virol J ; 8: 34, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21255440

RESUMO

BACKGROUND: The mechanism by which HIV infection leads to a selective depletion of CD4 cells leading to immunodeficiency remains highly debated. Whether the loss of CD4 cells is a direct consequence of virus infection or bystander apoptosis of uninfected cells is also uncertain. RESULTS: We have addressed this issue in the humanized mouse model of HIV infection using a HIV variant with a point mutation in the gp41 region of the Env glycoprotein that alters its fusogenic activity. We demonstrate here that a single amino acid change (V38E) altering the cell-to-cell fusion activity of the Env minimizes CD4 loss in humanized mice without altering viral replication. This differential pathogenesis was associated with a lack of bystander apoptosis induction by V38E virus even in the presence of similar levels of infected cells. Interestingly, immune activation was observed with both WT and V38E infection suggesting that the two phenomena are likely not interdependent in the mouse model. CONCLUSIONS: We conclude that Env fusion activity is one of the determinants of HIV pathogenesis and it may be possible to attenuate HIV by targeting gp41.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/virologia , Proteína gp41 do Envelope de HIV/metabolismo , HIV-1/patogenicidade , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Fatores de Virulência/metabolismo , Animais , Contagem de Linfócito CD4 , Proteína gp41 do Envelope de HIV/genética , Humanos , Camundongos , Camundongos SCID , Proteínas Mutantes/genética , Virulência , Fatores de Virulência/genética
12.
Nat Med ; 9(3): 347-51, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12579197

RESUMO

RNA interference (RNAi) is a powerful tool to silence gene expression post-transcriptionally. However, its potential to treat or prevent disease remains unproven. Fas-mediated apoptosis is implicated in a broad spectrum of liver diseases, where inhibiting hepatocyte death is life-saving. We investigated the in vivo silencing effect of small interfering RNA (siRNA) duplexes targeting the gene Fas (also known as Tnfrsf6), encoding the Fas receptor, to protect mice from liver failure and fibrosis in two models of autoimmune hepatitis. Intravenous injection of Fas siRNA specifically reduced Fas mRNA levels and expression of Fas protein in mouse hepatocytes, and the effects persisted without diminution for 10 days. Hepatocytes isolated from mice treated with Fas siRNA were resistant to apoptosis when exposed to Fas-specific antibody or co-cultured with concanavalin A (ConA)-stimulated hepatic mononuclear cells. Treatment with Fas siRNA 2 days before ConA challenge abrogated hepatocyte necrosis and inflammatory infiltration and markedly reduced serum concentrations of transaminases. Administering Fas siRNA beginning one week after initiating weekly ConA injections protected mice from liver fibrosis. In a more fulminant hepatitis induced by injecting agonistic Fas-specific antibody, 82% of mice treated with siRNA that effectively silenced Fas survived for 10 days of observation, whereas all control mice died within 3 days. Silencing Fas expression with RNAi holds therapeutic promise to prevent liver injury by protecting hepatocytes from cytotoxicity.


Assuntos
Hepatite Autoimune/prevenção & controle , Hepatite Autoimune/terapia , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Receptor fas/genética , Animais , Apoptose , Células Cultivadas , Concanavalina A/farmacologia , Modelos Animais de Doenças , Inativação Gênica , Hepatite Autoimune/genética , Hepatite Autoimune/patologia , Hepatócitos/patologia , Hepatócitos/fisiologia , Marcação In Situ das Extremidades Cortadas , Fígado/patologia , Masculino , Camundongos , Necrose , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor fas/metabolismo
13.
Nat Med ; 8(7): 681-6, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12042777

RESUMO

RNA interference silences gene expression through short interfering 21 23-mer double-strand RNA segments that guide mRNA degradation in a sequence-specific fashion. Here we report that siRNAs inhibit virus production by targeting the mRNAs for either the HIV-1 cellular receptor CD4, the viral structural Gag protein or green fluorescence protein substituted for the Nef regulatory protein. siRNAs effectively inhibit pre- and/or post-integration infection events in the HIV-1 life cycle. Thus, siRNAs may have potential for therapeutic intervention in HIV-1 and other viral infections.


Assuntos
Inativação Gênica , HIV-1/fisiologia , RNA não Traduzido/fisiologia , Sequência de Bases , Antígenos CD4/fisiologia , Linhagem Celular , HIV-1/efeitos dos fármacos , HIV-1/genética , Células HeLa , Humanos , RNA Antissenso/farmacologia , RNA Mensageiro/genética , RNA Interferente Pequeno , RNA não Traduzido/farmacologia , Receptores CCR5/fisiologia , Transfecção , Integração Viral/efeitos dos fármacos
14.
Mol Ther ; 18(5): 993-1001, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20216529

RESUMO

Inflammation mediated by tumor necrosis factor-alpha (TNF-alpha) and the associated neuronal apoptosis characterizes a number of neurologic disorders. Macrophages and microglial cells are believed to be the major source of TNF-alpha in the central nervous system (CNS). Here, we show that suppression of TNF-alpha by targeted delivery of small interfering RNA (siRNA) to macrophage/microglial cells dramatically reduces lipopolysaccharide (LPS)-induced neuroinflammation and neuronal apoptosis in vivo. Because macrophage/microglia express the nicotinic acetylcholine receptor (AchR) on their surface, we used a short AchR-binding peptide derived from the rabies virus glycoprotein (RVG) as a targeting ligand. This peptide was fused to nona-D-arginine residues (RVG-9dR) to enable siRNA binding. RVG-9dR was able to deliver siRNA to induce gene silencing in macrophages and microglia cells from wild type, but not AchR-deficient mice, confirming targeting specificity. Treatment with anti-TNF-alpha siRNA complexed to RVG-9dR achieved efficient silencing of LPS-induced TNF-alpha production by primary macrophages and microglia cells in vitro. Moreover, intravenous injection with RVG-9dR-complexed siRNA in mice reduced the LPS-induced TNF-alpha levels in blood as well as in the brain, leading to a significant reduction in neuronal apoptosis. These results demonstrate that RVG-9dR provides a tool for siRNA delivery to macrophages and microglia and that suppression of TNF-alpha can potentially be used to suppress neuroinflammation in vivo.


Assuntos
Macrófagos/metabolismo , RNA Interferente Pequeno/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Citometria de Fluxo , Inativação Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
Mol Ther ; 18(11): 2028-37, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20648001

RESUMO

Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in the activation of T cells. RNA interference (RNAi)-mediated silencing of negative immunoregulatory molecules expressed by DCs may provide a strategy to enhance the potency of DC-based vaccines and immunotherapy. Ablation of suppressor of cytokine signaling-1 (SOCS-1) in antigen-presenting cells has been shown to enhance cellular immune response in mice. Here, we used a previously reported DC-targeting approach to deliver small interfering RNA (siRNA) against SOCS-1 to human myeloid-derived DCs (MDDCs). SOCS1-silencing in MDDCs resulted in enhanced cytokine responses to lipopolysaccharide (LPS) and a strong mixed-lymphocyte reaction. Moreover, only DCs treated with SOCS-1 siRNA, and not controls, elicited strong primary in vitro responses to well-characterized HLA-A*0201-restricted Melan-A/MART-1 and human immunodeficiency virus (HIV) Gag epitopes in naive CD8(+) T cells from healthy donors. Finally, stimulation of CD8(+) T cells from HIV-seropositive subjects with SOCS1-silenced DCs resulted in an augmented polyfunctional cytotoxic T-lymphocyte (CTL) response, suggesting that SOCS-1 silencing can restore functionally compromised T cells in HIV infection. Collectively, these results demonstrate the feasibility of DC3-9dR-mediated manipulation of DC function to enhance DC immunogenicity for potential vaccine or immunotherapeutic applications.


Assuntos
Células Dendríticas/imunologia , HIV/imunologia , RNA Interferente Pequeno/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Apoptose , Western Blotting , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Citometria de Fluxo , HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Humanos , Lipopolissacarídeos/farmacologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Fragmentos de Peptídeos/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/antagonistas & inibidores , Proteínas Supressoras da Sinalização de Citocina/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
16.
Mol Ther ; 18(2): 370-6, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19997090

RESUMO

RNA interference (RNAi)-mediated knockdown of gene expression offers a novel treatment strategy for human immunodeficiency virus (HIV) infection. However, the major hurdle for clinical use is a practical strategy for small interfering RNA (siRNA) delivery to the multiple immune cell types important in viral pathogenesis. We have developed a novel immunoliposome method targeting the lymphocyte function-associated antigen-1 (LFA-1) integrin expressed on all leukocytes and evaluated it for systemic delivery of siRNA in a humanized mouse model. We show that in vivo administration of the LFA-1 integrin-targeted and stabilized nanoparticles (LFA-1 I-tsNPs) results in selective uptake of siRNA by T cells and macrophages, the prime targets of HIV. Further, in vivo administration of anti-CCR5 siRNA/LFA-1 I-tsNPs resulted in leukocyte-specific gene silencing that was sustained for 10 days. Finally, humanized mice challenged with HIV after anti-CCR5 siRNA treatment showed enhanced resistance to infection as assessed by the reduction in plasma viral load and disease-associated CD4 T-cell loss. This study demonstrates the potential in vivo applicability of LFA-1-directed siRNA delivery as anti-HIV prophylaxis.


Assuntos
Inativação Gênica/fisiologia , Infecções por HIV/prevenção & controle , Antígeno-1 Associado à Função Linfocitária/fisiologia , Nanopartículas/uso terapêutico , RNA Interferente Pequeno/fisiologia , Receptores CCR5/genética , Animais , Infecções por HIV/genética , Infecções por HIV/imunologia , Leucócitos/metabolismo , Lipossomos/uso terapêutico , Antígeno-1 Associado à Função Linfocitária/genética , Camundongos , Interferência de RNA , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Biochem Biophys Res Commun ; 374(2): 214-8, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18619945

RESUMO

The high genetic diversity and mutability of HIV pose a major problem for RNAi-mediated antiviral therapy. Simultaneous targeting of multiple highly conserved viral sequences has been suggested for durable cross-clade inhibition. Here we validate the approach of co-targeting two conserved sequences in the Tat and Vif genes. When coexpressed as artificial microRNA from a PolII driven miR-155-based vector, the sequences together mediated effective and sustained inhibition of HIV replication without virus breakout. To understand the nature of this efficient control, we analyzed genome sequences of 625 HIV-1 isolates in the Los Alamos Sequence database. Interestingly most natural variants were capable of wobble binding with the Tat/Vif siRNAs. Efficient silencing of reporter luciferase constructs bearing these variants residues verified that the Tat/Vif sequences together tolerated wobble binding and mediated functional RNAi. We propose the rationale of targeting highly conserved HIV sequences where wobble substitutions permit functional RNAi for global HIV repression.


Assuntos
Sequência Conservada , HIV-1/fisiologia , MicroRNAs/genética , Replicação Viral , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Pareamento de Bases , Linhagem Celular , Inativação Gênica , Genes Reporter , Vetores Genéticos , HIV-1/genética , Humanos , Replicação Viral/genética
18.
PLoS Pathog ; 2(12): e130, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17173480

RESUMO

To replicate, HIV-1 capitalizes on endogenous cellular activation pathways resulting in recruitment of key host transcription factors to its viral enhancer. RNA interference has been a powerful tool for blocking key checkpoints in HIV-1 entry into cells. Here we apply RNA interference to HIV-1 transcription in primary macrophages, a major reservoir of the virus, and specifically target the transcription factor NFAT5 (nuclear factor of activated T cells 5), which is the most evolutionarily divergent NFAT protein. By molecularly cloning and sequencing isolates from multiple viral subtypes, and performing DNase I footprinting, electrophoretic mobility shift, and promoter mutagenesis transfection assays, we demonstrate that NFAT5 functionally interacts with a specific enhancer binding site conserved in HIV-1, HIV-2, and multiple simian immunodeficiency viruses. Using small interfering RNA to ablate expression of endogenous NFAT5 protein, we show that the replication of three major HIV-1 viral subtypes (B, C, and E) is dependent upon NFAT5 in human primary differentiated macrophages. Our results define a novel host factor-viral enhancer interaction that reveals a new regulatory role for NFAT5 and defines a functional DNA motif conserved across HIV-1 subtypes and representative simian immunodeficiency viruses. Inhibition of the NFAT5-LTR interaction may thus present a novel therapeutic target to suppress HIV-1 replication and progression of AIDS.


Assuntos
HIV-1/fisiologia , Macrófagos/virologia , Interferência de RNA , Sequências Repetidas Terminais/fisiologia , Fatores de Transcrição/fisiologia , Replicação Viral/fisiologia , Síndrome da Imunodeficiência Adquirida/fisiopatologia , Linhagem Celular , Células Cultivadas , DNA Viral , Progressão da Doença , Regulação da Expressão Gênica , HIV-1/genética , HIV-1/patogenicidade , Células HeLa , Humanos , Macrófagos/fisiologia , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sequências Repetidas Terminais/genética , Fatores de Transcrição/genética , Replicação Viral/genética
19.
Nat Biotechnol ; 23(6): 709-17, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15908939

RESUMO

Delivery of small interfering RNAs (siRNAs) into cells is a key obstacle to their therapeutic application. We designed a protamine-antibody fusion protein to deliver siRNA to HIV-infected or envelope-transfected cells. The fusion protein (F105-P) was designed with the protamine coding sequence linked to the C terminus of the heavy chain Fab fragment of an HIV-1 envelope antibody. siRNAs bound to F105-P induced silencing only in cells expressing HIV-1 envelope. Additionally, siRNAs targeted against the HIV-1 capsid gene gag, inhibited HIV replication in hard-to-transfect, HIV-infected primary T cells. Intratumoral or intravenous injection of F105-P-complexed siRNAs into mice targeted HIV envelope-expressing B16 melanoma cells, but not normal tissue or envelope-negative B16 cells; injection of F105-P with siRNAs targeting c-myc, MDM2 and VEGF inhibited envelope-expressing subcutaneous B16 tumors. Furthermore, an ErbB2 single-chain antibody fused with protamine delivered siRNAs specifically into ErbB2-expressing cancer cells. This study demonstrates the potential for systemic, cell-type specific, antibody-mediated siRNA delivery.


Assuntos
Sistemas de Liberação de Medicamentos , RNA Interferente Pequeno/administração & dosagem , Receptores de Superfície Celular/fisiologia , Animais , Anticorpos/química , Células COS , HIV-1 , Humanos , Interferons/metabolismo , Melanoma Experimental/terapia , Camundongos , Receptores de HIV/fisiologia , Proteínas Recombinantes de Fusão
20.
Heliyon ; 4(2): e00545, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29527580

RESUMO

We have previously reported that overexpression of Programmed Death -1 Homolog (PD-1H) in human monocytes leads to activation and spontaneous secretion of multiple pro inflammatory cytokines. Here we evaluate changes in monocytes gene expression after enforced PD-1H expression by gene array. The results show that there are significant alterations in 51 potential candidate genes that relate to immune response, cell adhesion and metabolism. Genes corresponding to pro-inflammatory cytokines showed the highest upregulation, 7, 3.2, 3.0, 5.8, 4.4 and 3.1 fold upregulation of TNF-α, IL-1 ß, IFN-α, γ, λ and IL-27 relative to vector control. The data are in agreement with cytometric bead array analysis showing induction of proinflammatory cytokines, IL-6, IL-1ß and TNF-α by PD-1H. Other genes related to inflammation, include transglutaminase 2 (TG2), NF-κB (p65 and p50) and toll like receptors (TLR) 3 and 4 were upregulated 5, 4.5 and 2.5 fold, respectively. Gene set enrichment analysis (GSEA) also revealed that signaling pathways related to inflammatory response, such as NFκB, AT1R, PYK2, MAPK, RELA, TNFR1, MTOR and proteasomal degradation, were significantly upregulated in response to PD-1H overexpression. We validated the results utilizing a standard inflammatory sepsis model in humanized BLT mice, finding that PD-1H expression was highly correlated with proinflammatory cytokine production. We therefore conclude that PD-1H functions to enhance monocyte activation and the induction of a pro-inflammatory gene expression profile.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA