Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Immunol ; 17(8): 906-13, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27434011

RESUMO

Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1ß are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1ß, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.


Assuntos
Inflamação/fisiopatologia , Interleucina-1alfa/fisiologia , Alarminas/metabolismo , Animais , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Granuloma/etiologia , Granuloma/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-1alfa/biossíntese , Interleucina-1alfa/genética , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Ligação Proteica , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Receptores de Interleucina-1/fisiologia , Transdução de Sinais
2.
Mol Ther ; 32(1): 103-123, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37919899

RESUMO

Targeted delivery and cell-type-specific expression of gene-editing proteins in various cell types in vivo represent major challenges for all viral and non-viral delivery platforms developed to date. Here, we describe the development and analysis of artificial vectors for intravascular delivery (AVIDs), an engineered adenovirus-based gene delivery platform that allows for highly targeted, safe, and efficient gene delivery to human hematopoietic stem and progenitor cells (HSPCs) in vivo after intravenous vector administration. Due to a set of refined structural modifications, intravenous administration of AVIDs did not trigger cytokine storm, hepatotoxicity, or thrombocytopenia. Single intravenous administration of AVIDs to humanized mice, grafted with human CD34+ cells, led to up to 20% transduction of CD34+CD38-CD45RA- HSPC subsets in the bone marrow. Importantly, targeted in vivo transduction of CD34+CD38-CD45RA-CD90-CD49f+ subsets, highly enriched for human hematopoietic stem cells (HSCs), reached up to 19%, which represented a 1,900-fold selectivity in gene delivery to HSC-enriched over lineage-committed CD34-negative cell populations. Because the AVID platform allows for regulated, cell-type-specific expression of gene-editing technologies as well as expression of immunomodulatory proteins to ensure persistence of corrected HSCs in vivo, the HSC-targeted AVID platform may enable development of curative therapies through in vivo gene correction in human HSCs after a single intravenous administration.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Animais , Camundongos , Células-Tronco Hematopoéticas/metabolismo , Técnicas de Transferência de Genes , Antígenos CD34/metabolismo , Terapia Genética , Adenoviridae/genética , Adenoviridae/metabolismo
3.
Immunity ; 43(6): 1125-36, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26682985

RESUMO

The interleukin-1 receptor I (IL-1RI) is critical for host resistance to Mycobacterium tuberculosis (Mtb), yet the mechanisms of IL-1RI-mediated pathogen control remain unclear. Here, we show that without IL-1RI, Mtb-infected newly recruited Ly6G(hi) myeloid cells failed to upregulate tumor necrosis factor receptor I (TNF-RI) and to produce reactive oxygen species, resulting in compromised pathogen control. Furthermore, simultaneous ablation of IL-1RI and TNF-RI signaling on either stroma or hematopoietic cells led to early lethality, indicating non-redundant and synergistic roles of IL-1 and TNF in mediating macrophage-stroma cross-talk that was critical for optimal control of Mtb infection. Finally, we show that even in the presence of functional Mtb-specific adaptive immunity, the lack of IL-1α and not IL-1ß led to an exuberant intracellular pathogen replication and progressive non-resolving inflammation. Our study reveals functional interdependence between IL-1 and TNF in enabling Mtb control mechanisms that are critical for host survival.


Assuntos
Interleucina-1alfa/imunologia , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Separação Celular , Modelos Animais de Doenças , Citometria de Fluxo , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis , Receptores Tipo I de Interleucina-1/imunologia
4.
Immunity ; 38(2): 203-5, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23438818

RESUMO

The molecular mechanisms that regulate functional activation of IL-1α remain elusive. In this issue of Immunity, Zheng et al. (2013) describe a molecular system implicating interleukin-1 receptor-2 (IL-1R2) as a principal cytosolic factor that controls functional IL-1α activation during necrosis.

5.
Immunity ; 31(1): 110-21, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19576795

RESUMO

The recognition of viral components by host pattern-recognition receptors triggers the induction of the antiviral innate immune response. Toll-like receptor 9 (TLR9) and NLRP3 inflammasome were shown to be the principal specific sensors of viral double-stranded DNA. Here we present evidence that macrophages in vivo activated an innate immune response to a double-stranded DNA virus, adenovirus (Ad), independently of TLR9 or NLRP3 inflammasome. In response to Ad, macrophage-derived IL-1 alpha triggered IL-1RI-dependent production of a defined set of proinflammatory cytokines and chemokines. The IL-1 alpha-mediated response required a selective interaction of virus arginine-glycine-aspartic acid (RGD) motifs with macrophage beta(3) integrins. Thus, these data identify IL-1 alpha-IL-1RI as a key pathway allowing for the activation of proinflammatory responses to the virus, independently of its genomic nucleic acid recognition.


Assuntos
Proteínas de Transporte/imunologia , Interleucina-1alfa/imunologia , Macrófagos/imunologia , Receptores de Interleucina-1/imunologia , Receptor Toll-Like 9/imunologia , Adenoviridae/imunologia , Adenoviridae/metabolismo , Animais , Proteínas de Transporte/metabolismo , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Imunidade Inata , Integrina beta3/imunologia , Integrina beta3/metabolismo , Interleucina-1alfa/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores de Interleucina-1/metabolismo , Baço/citologia , Baço/imunologia , Baço/metabolismo , Receptor Toll-Like 9/metabolismo
6.
PLoS Pathog ; 10(3): e1004035, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651866

RESUMO

Inflammation is a highly coordinated host response to infection, injury, or cell stress. In most instances, the inflammatory response is pro-survival and is aimed at restoring physiological tissue homeostasis and eliminating invading pathogens, although exuberant inflammation can lead to tissue damage and death. Intravascular injection of adenovirus (Ad) results in virus accumulation in resident tissue macrophages that trigger activation of CXCL1 and CXCL2 chemokines via the IL-1α-IL-1RI signaling pathway. However, the mechanistic role and functional significance of this pathway in orchestrating cellular inflammatory responses to the virus in vivo remain unclear. Resident metallophilic macrophages expressing macrophage receptor with collagenous structure (MARCO+) in the splenic marginal zone (MZ) play the principal role in trapping Ad from the blood. Here we show that intravascular Ad administration leads to the rapid recruitment of Ly-6G+7/4+ polymorphonuclear leukocytes (PMNs) in the splenic MZ, the anatomical compartment that remains free of PMNs when these cells are purged from the bone marrow via a non-inflammatory stimulus. Furthermore, PMN recruitment in the splenic MZ resulted in elimination of virus-containing cells. IL-1α-IL-1RI signaling is only partially responsible for PMN recruitment in the MZ and requires CXCR2, but not CXCR1 signaling. We further found reduced recruitment of PMNs in the splenic MZ in complement C3-deficient mice, and that pre-treatment of IL-1α-deficient, but not wild-type mice, with complement inhibitor CR2-Crry (inhibits all complement pathways at C3 activation) or CR2-fH (inhibits only the alternative complement activation pathway) prior to Ad infection, abrogates PMN recruitment to the MZ and prevents elimination of MARCO+ macrophages from the spleen. Collectively, our study reveals a non-redundant role of the molecular factors of innate immunity--the chemokine-activating IL-1α-IL-1RI-CXCR2 axis and complement--in orchestrating local inflammation and functional cooperation of PMNs and resident macrophages in the splenic MZ, which collectively contribute to limiting disseminated pathogen spread via elimination of virus-containing cells.


Assuntos
Infecções por Adenoviridae/imunologia , Proteínas do Sistema Complemento/imunologia , Inflamação/imunologia , Interleucina-1alfa/imunologia , Neutrófilos/imunologia , Adenoviridae/imunologia , Animais , Quimiocinas/metabolismo , Quimiotaxia de Leucócito/imunologia , Citometria de Fluxo , Imunofluorescência , Imuno-Histoquímica , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Baço/citologia , Baço/imunologia
7.
Nature ; 450(7166): 110-4, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17934446

RESUMO

Lymph nodes prevent the systemic dissemination of pathogens such as viruses that infect peripheral tissues after penetrating the body's surface barriers. They are also the staging ground of adaptive immune responses to pathogen-derived antigens. It is unclear how virus particles are cleared from afferent lymph and presented to cognate B cells to induce antibody responses. Here we identify a population of CD11b+CD169+MHCII+ macrophages on the floor of the subcapsular sinus (SCS) and in the medulla of lymph nodes that capture viral particles within minutes after subcutaneous injection. Macrophages in the SCS translocated surface-bound viral particles across the SCS floor and presented them to migrating B cells in the underlying follicles. Selective depletion of these macrophages compromised local viral retention, exacerbated viraemia of the host, and impaired local B-cell activation. These findings indicate that CD169+ macrophages have a dual physiological function. They act as innate 'flypaper' by preventing the systemic spread of lymph-borne pathogens and as critical gatekeepers at the lymph-tissue interface that facilitate the recognition of particulate antigens by B cells and initiate humoral immune responses.


Assuntos
Linfócitos B/imunologia , Linfonodos/imunologia , Linfonodos/virologia , Linfa/virologia , Macrófagos/imunologia , Vesiculovirus/imunologia , Animais , Formação de Anticorpos/imunologia , Antígeno CD11b/metabolismo , Linhagem Celular , Linfonodos/citologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores Imunológicos/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico
8.
Mol Ther ; 18(8): 1422-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20551916

RESUMO

The innate immune and inflammatory response represents one of the key stumbling blocks limiting the efficacy of viral-based therapies. Numerous human diseases could be corrected or ameliorated if viruses were harnessed to safely and effectively deliver therapeutic genes to diseased cells and tissues in vivo. Recent studies have shown that host cells recognize viruses using an elaborate network of sensor proteins localized at the plasma membrane, in endosomes, or in the cytosol. Three classes of sensors have been implicated in sensing viruses in mammalian cells-Toll-like receptors (TLRs), retinoid acid-inducible gene (RIG)-I-like receptors (RLRs), and nucleotide oligomerization domain (NOD)-like receptors (NLRs). The interaction of virus-associated nucleic acids with these sensor molecules triggers a signaling cascade that activates the principal host defense program aimed to limit or eliminate virus infection and restore tissue homeostasis. In addition, recent data strongly suggest that host cells can mount innate immune responses to viruses without prior recognition of their nucleic acids. To deliver therapeutic genes into the nuclei of diseased cells, viral gene therapy vectors must be efficient at penetrating either the plasma or endosomal membrane. The therapeutic use of high numbers of virus particles disturbs cellular homeostasis, triggering cell damage and stress pathways, or "sensing of modified self". Accumulating data indicate that the sensing of modified self might represent a powerful framework explaining the innate immune response activation by viral gene therapy vectors.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Vírus/genética , Animais , Humanos , Modelos Biológicos , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
9.
Mol Ther ; 17(4): 675-84, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19223863

RESUMO

Human adenovirus (Ad) is a ubiquitous pathogen causing a wide range of diseases. Although the interactions of human Ad serotype 5 (Ad5) with susceptible cells in vitro are known in great detail, host factors controlling the tissue specificity of Ad5 infection in vivo remain poorly understood. Here, we analyzed the mechanisms of sequestration by the liver for blood-born human Ads and Ad5-based vectors. Our data suggest that several known mechanisms that lead to Ad5 sequestration by the liver become engaged in a redundant, sequential, and synergistic manner to ensure the rapid clearance of circulating virus particles from the blood. These mechanisms include (i) trapping of the virus by liver residential macrophages, Kupffer cells; (ii) Ad5 hepatocyte infection via blood factor-hexon interactions; and (iii) Ad5 penton RGD motif-mediated interactions with liver endothelial cells and hepatocytes, mediating virus retention in the space of Disse. More important, we show that when all of these mechanisms are simultaneously inactivated via mutations of Ad5 capsid proteins and pharmacological interventions, virus sequestration by the liver is markedly reduced. Therefore, our study is the first demonstration of the principal possibility of ablating the sequestration of blood-born Ad in the liver via specific inactivation of a defined set of mechanisms that control this process.


Assuntos
Adenoviridae/isolamento & purificação , Patógenos Transmitidos pelo Sangue , Fígado/virologia , Adenoviridae/genética , Animais , Proteínas do Capsídeo/metabolismo , Fator X/metabolismo , Humanos , Células de Kupffer/patologia , Fígado/patologia , Camundongos , Mutação , Oligopeptídeos/metabolismo , Ligação Proteica , Transdução Genética , Proteínas Virais/genética
10.
Biomaterials ; 225: 119512, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31585233

RESUMO

For decades, cationic polymer nanoparticles have been investigated for nucleic acid delivery. Despite promising in vitro transfection results, most formulations have failed to translate into the clinic due to significant in vivo toxicity - especially when delivered intravenously. To address this significant problem, we investigated the detailed mechanisms that govern the complex in vivo systemic toxicity response to common polymeric nanoparticles. We determined that the toxicity response is material dependent. For branched polyethylenimine (bPEI) nanoparticles - toxicity is a function of multiple pathophysiological responses - triggering of innate immune sensors, induction of hepatic toxicity, and significant alteration of hematological properties. In contrast, for chitosan-based nanoparticles - systemic toxicity is primarily driven through innate immune activation. We further identified that modification of primary amines to secondary and tertiary amines using the small molecule imidazole-acetic-acid (IAA) ameliorates in vivo toxicity from both nanocarriers by different, material-specific mechanisms related to Toll-like receptor 4 activation (for bPEI) and complement activation driven neutrophil infiltration (for chitosan), respectively. Our results provide a detailed roadmap for evaluating in vivo toxicity of nanocarriers and identifies potential opportunities to reduce toxicity for eventual clinical translation.


Assuntos
Aminas/química , Proteínas do Sistema Complemento/metabolismo , Nanopartículas/toxicidade , Receptor 4 Toll-Like/metabolismo , Ácido Acético/química , Animais , Cátions , Quitosana/química , Quitosana/toxicidade , Feminino , Imidazóis/química , Camundongos , Camundongos Endogâmicos C57BL , Polietilenoimina/química , Polietilenoimina/toxicidade , Coroa de Proteína/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo
11.
Cancer Res ; 66(2): 960-9, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424031

RESUMO

Heat shock proteins such as gp96 have the ability to chaperone peptides and activate antigen-presenting cells. In this study, we tested whether adenovirus-mediated overexpression of secreted or membrane-associated forms of gp96 in tumor cells would stimulate an antitumor immune response. Studies were carried out in C57Bl/6 mice bearing aggressively growing s.c. tumors derived from syngeneic TC-1 cells, a cell line that expresses HPV16 E6 and E7 proteins. We found that secreted gp96 can induce protective and therapeutic antitumor immune responses. Our data also indicate that the antitumor effect of sgp96 expression seems to be limited by the induction of suppressive regulatory T cells (Treg). TC-1 tumor transplantation increased the number of splenic and tumor-infiltrating Tregs. Importantly, treatment of mice with low-dose cyclophosphamide decreased the number of Tregs and enhanced the immunostimulatory effect of sgp96 expression. We also tested whether an oncolytic vector (Ad.IR-E1A/TRAIL), that is able to induce tumor cell apoptosis and, potentially, release cryptic tumor epitopes in immunogenic form, could stimulate antitumor immune responses. Although tumor cells infected ex vivo with Ad.IR-E1A/TRAIL had no antitumor effect when used as a vaccine alone, the additional treatment with low-dose cyclophosphamide resulted in the elimination of pre-established tumors. This study gives a rationale for testing approaches that suppress Tregs in combination with oncolytic or immunostimulatory vectors.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Ciclofosfamida/farmacologia , Proteínas de Choque Térmico/biossíntese , Adenoviridae/genética , Animais , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer , Relação Dose-Resposta a Droga , Vetores Genéticos , Papillomavirus Humano 16 , Humanos , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Neoplasias/terapia , Proteínas Oncogênicas Virais , Terapia Viral Oncolítica , Proteínas Repressoras , Transplante Heterólogo , Células Tumorais Cultivadas/imunologia
12.
Front Immunol ; 5: 353, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25101093

RESUMO

Human oncogenic viruses include Epstein-Barr virus, hepatitis B virus, hepatitis C virus, human papilloma virus, human T-cell lymphotropic virus, Kaposi's associated sarcoma virus, and Merkel cell polyomavirus. It would be expected that during virus-host interaction, the immune system would recognize these pathogens and eliminate them. However, through evolution, these viruses have developed a number of strategies to avoid such an outcome and successfully establish chronic infections. The persistent nature of the infection caused by these viruses is associated with their oncogenic potential. In this article, we will review the latest information on the interaction between oncogenic viruses and the innate immune system of the host. In particular, we will summarize the available knowledge on the recognition by host pattern-recognition receptors of pathogen-associated molecular patterns present in the incoming viral particle or generated during the virus' life cycle. We will also review the data on the recognition of cell-derived danger associated molecular patterns generated during the virus infection that may impact the outcome of the host-pathogen interaction and the development cancer.

13.
Methods Mol Biol ; 1089: 133-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24132483

RESUMO

Even though natural infections with adenovirus (Ad) are largely harmless in humans, an intravenous Ad vector administration for gene delivery purposes, especially at high doses, stimulates strong innate and adaptive immune responses, and can be fatal to the host. In animal models, intravenous Ad administration has been shown to induce transcription and release in the serum of a great number of pro-inflammatory cytokines and chemokines. Macrophages, including tissue residential macrophages (e.g., Kupffer cells in the liver), and dendritic cells throughout the body are considered to be the primary source of these pro-inflammatory mediators following their transduction with Ads. Here, we provide an overview and methodology for the qualitative and quantitative analyses of pro-inflammatory cytokine and chemokine expression in the spleen and their release into the bloodstream after intravenous Ad delivery using antibody arrays.


Assuntos
Adenoviridae/imunologia , Anticorpos Antivirais , Imunidade Inata , Análise Serial de Proteínas/métodos , Animais , Quimiocinas/sangue , Quimiocinas/imunologia , Citocinas/sangue , Citocinas/imunologia , Humanos , Mediadores da Inflamação/sangue , Mediadores da Inflamação/imunologia , Camundongos , Baço/imunologia , Baço/virologia
14.
Cell Rep ; 3(6): 1840-6, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23770239

RESUMO

Although molecular components that execute noninflammatory apoptotic cell death are well defined, molecular pathways that trigger necrotic cell death remain poorly characterized. Here, we show that in response to infection with adenovirus or Listeria monocytogenes, macrophages in vivo undergo rapid proinflammatory necrotic death that is controlled by interferon-regulatory factor 3 (IRF3). The transcriptional activity of IRF3 is, surprisingly, not required for the induction of necrosis, and it proceeds normally in mice deficient in all known regulators of necrotic death or IRF3 activation, including RIPK3, caspases 1, 8, or 11, STING, and IPS1/MAVS. Although L. monocytogenes triggers necrosis to promote the infection, IRF3-dependent necrosis is required for reducing pathogen burden in the models of disseminated infection with adenovirus. Therefore, our studies implicate IRF3 as a principal and nonredundant component of a physiologically regulated necrotic cell-death pathway that operates as an effective innate immune mechanism of host protection against disseminated virus infection.


Assuntos
Infecções por Adenovirus Humanos/imunologia , Adenovírus Humanos/imunologia , Fator Regulador 3 de Interferon/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Infecções por Adenovirus Humanos/patologia , Animais , Caspases/metabolismo , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Listeriose/patologia , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/imunologia , Necrose/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais
15.
Science ; 338(6108): 795-8, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23019612

RESUMO

Although coagulation factors play a role in host defense for "living fossils" such as horseshoe crabs, the role of the coagulation system in immunity in higher organisms remains unclear. We modeled the interface of human species C adenovirus (HAdv) interaction with coagulation factor X (FX) and introduced a mutation that abrogated formation of the HAdv-FX complex. In vivo genome-wide transcriptional profiling revealed that FX-binding-ablated virus failed to activate a distinct network of nuclear factor κB-dependent early-response genes that are activated by HAdv-FX complex downstream of TLR4/MyD88/TRIF/TRAF6 signaling. Our study implicates host factor "decoration" of the virus as a mechanism to trigger an innate immune sensor that responds to a misplacement of coagulation FX from the blood into intracellular macrophage compartments upon virus entry into the cell.


Assuntos
Infecções por Adenoviridae/imunologia , Adenovírus Humanos/imunologia , Adenovírus Humanos/metabolismo , Fator X/metabolismo , Imunidade Inata , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Adenovírus Humanos/genética , Animais , Células CHO , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Microscopia Crioeletrônica , Citocinas/metabolismo , Fator X/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hepatócitos/virologia , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Dinâmica Molecular , Mutação , NF-kappa B/metabolismo , Transdução de Sinais , Internalização do Vírus
17.
Curr Opin Mol Ther ; 11(5): 523-31, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19806500

RESUMO

Adenovirus (Ad) vectors have substantial potential as biological therapeutics for the treatment of human diseases. Evidence from preclinical studies and clinical trials indicated that several acquired and inherited diseases could be corrected or ameliorated with cell type-specific Ad targeting. One of the major barriers for in vivo Ad targeting is the sequestration of the blood-borne virus in the liver. Significant recent advances have been made in understanding the molecular mechanisms involved in mediating Ad sequestration in the liver. Recognizing the redundancy and synergism between the mechanisms that mediate Ad liver cell transduction and those that mediate the sequestration of blood-borne Ads in the liver creates an opportunity for the development of safe and targeted Ads for gene therapy applications.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Fígado/virologia , Animais , Vetores Genéticos/genética , Hepatócitos/virologia , Humanos , Células de Kupffer/virologia , Fígado/patologia , Modelos Biológicos , Transdução Genética
18.
J Virol ; 81(22): 12249-59, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17855526

RESUMO

The molecular mechanisms governing the infectivity of adenovirus (Ad) toward specific cell and tissue types in vivo remain poorly understood. The direct Ad binding to hepatic heparan sulfate proteoglycans via the KKTK motif within the fiber shaft domain was suggested to be the major mechanism of Ad liver cell infection in vivo. Here, we describe the generation and in vitro and in vivo infectivity studies of Ad5-based vectors possessing long Ad31- or Ad41-derived fiber shaft domains, which lack the KKTK motif. We found that all the critical early steps of Ad infection, including attachment to the cellular receptor, internalization, and virus genome transfer into the nucleus, occurred with similar levels of efficiency for fiber shaft-chimeric vectors and unmodified Ad5. Upon intravenous delivery into mice, fiber shaft-chimeric vectors accumulated in liver tissue, transduced liver cells, and induced the production of proinflammatory cytokines (tumor necrosis factor alpha and interleukin-6) and the chemokine monocyte chemoattractant protein 1 at levels indistinguishable from those observed for Ad5. Thus, our data provide evidence that the Ad5 fiber shaft amino acid sequence does not play any substantial role in determining adenovirus infectivity toward hepatic cells in vivo. The data obtained contribute to improving our understanding of the molecular mechanisms determining Ad infectivity and biodistribution in vivo and may aid in designing novel Ad-based vectors for gene therapy applications.


Assuntos
Adenoviridae/fisiologia , Proteínas do Capsídeo/genética , Vetores Genéticos/fisiologia , Hepatócitos/metabolismo , Adenoviridae/genética , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Linhagem Celular Tumoral , Vetores Genéticos/genética , Heparina/farmacologia , Hepatócitos/efeitos dos fármacos , Humanos , Células de Kupffer/metabolismo , Camundongos , Dados de Sequência Molecular , Transdução Genética
19.
Mol Ther ; 15(8): 1410-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17505469

RESUMO

Intravascular delivery of adenovirus (Ad) vectors is being developed for liver-directed gene therapy for targeting disseminated disease in cancer therapeutics and for targeting non-hepatic tissues and organs through vector engineering strategies. The utility of Ad vectors is not limited to serotype 5 (Ad5), and many alternate human serotypes and non-human serotypes of Ad are currently being investigated. Critical to intravascular delivery of Ad is the interaction of the virus with host blood cells and plasma proteins, because immediate contact is observed following injection. Although incompletely understood, recent studies suggest that these interactions are critical in dictating the particle bio-distribution and resulting transduction properties of Ad in vivo. For example, plasma proteins-in particular, vitamin K-dependent coagulation zymogens-are able to directly bind to Ad, and "bridge" the virus to receptors in the liver. Unraveling and characterizing these mechanisms will be of fundamental importance both for understanding basic Ad biology in vivo and for refinement and optimization of Ad vectors for human gene therapy.


Assuntos
Adenoviridae/fisiologia , Células Sanguíneas/virologia , Terapia Genética/métodos , Transdução Genética/métodos , Animais , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Humanos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA