Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
BMC Immunol ; 19(1): 26, 2018 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-30055563

RESUMEN

BACKGROUND: Agonists of TLR3 and TLR4 are effective immunoadjuvants for different types of vaccines. The mechanisms of their immunostimulatory action differ significantly; these differences are particularly critical for immunization with non-replicating adenovirus vectors (rAds) based vaccines. Unlike traditional vaccines, rAd based vaccines are not designed to capture vaccine antigens from the external environment by antigen presenting cells (APCs), but rather they are targeted to the de novo synthesis of vaccine antigens in APCs transfected with rAd. To date, there is no clear understanding about approaches to improve the efficacy of rAd vaccinations with immunoadjuvants. In this study, we investigated the immunoadjuvant effect of TLR3 and TLR4 agonists on the level of activation of APCs during vaccination with rAds. RESULTS: We demonstrated that TLR3 and TLR4 agonists confer different effects on the molecular processes in APCs that determine the efficacy of antigen delivery and activation of antigen-specific CD4+ and CD8+ T cells. APCs activated with agonists of TLR4 were characterized by up-regulated production of target antigen mRNA and protein encoded in rAd, as well as enhanced expression of the co-activation receptors CD80, CD86 and CD40, and pro-inflammatory cytokines TNF-α, IL6 and IL12. These effects of TLR4 agonists have provided a significant increase in the number of antigen-specific CD4+ and CD8+ T cells. TLR3 agonist, on the contrary, inhibited transcription and synthesis of rAd-encoded antigens, but improved expression of CD40 and IFN-ß in APCs. The cumulative effect of TLR3 agonist have resulted in only a slight improvement in the activation of antigen-specific T cells. Also, we demonstrated that IFN-ß and TNF-α, secreted by APCs in response to TLR3 and TLR4 agonists, respectively, have an opposite effect on the transcription of the targeted gene encoded in rAd. Specifically, IFN-ß inhibited, and TNF-α stimulated the expression of target vaccine antigens in APCs. CONCLUSIONS: Our data demonstrate that agonists of TLR4 but not TLR3 merit further study as adjuvants for development of vaccines based on recombinant adenoviral vectors.


Asunto(s)
Vacunas contra el Adenovirus/inmunología , Adyuvantes Inmunológicos/farmacología , Presentación de Antígeno , Células Dendríticas/efectos de los fármacos , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 4/agonistas , Animales , Antígeno B7-1/inmunología , Antígeno B7-2/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Vectores Genéticos/inmunología , Inmunización , Interferón beta/inmunología , Interleucina-12/inmunología , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Células TH1/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Vacunas Sintéticas/inmunología
2.
Infect Immun ; 81(10): 3855-64, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897616

RESUMEN

Pathogen recognition receptors (PRRs) are essential components of host innate immune systems that detect specific conserved pathogen-associated molecular patterns (PAMPs) presented by microorganisms. Members of two families of PRRs, transmembrane Toll-like receptors (TLRs 1, 2, 4, 5, and 6) and cytosolic NOD receptors (NOD1 and NOD2), are stimulated upon recognition of various bacterial PAMPs. Such stimulation leads to induction of a number of immune defense reactions, mainly triggered via activation of the transcription factor NF-κB. While coordination of responses initiated via different PRRs sensing multiple PAMPS present during an infection makes clear biological sense for the host, such interactions have not been fully characterized. Here, we demonstrate that combined stimulation of NOD1 and TLR5 (as well as other NOD and TLR family members) strongly potentiates activity of NF-κB and induces enhanced levels of innate immune reactions (e.g., cytokine production) both in vitro and in vivo. Moreover, we show that an increased level of NF-κB activity plays a critical role in formation of downstream responses. In live mice, synergy between these receptors resulting in potentiation of NF-κB activity was organ specific, being most prominent in the gastrointestinal tract. Coordinated activity of NOD1 and TLR5 significantly increased protection of mice against enteroinvasive Salmonella infection. Obtained results suggest that cooperation of NOD and TLR receptors is important for effective responses to microbial infection in vivo.


Asunto(s)
Inmunidad Innata/fisiología , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Salmonella typhimurium , Receptor Toll-Like 5/metabolismo , Animales , Femenino , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , Proteína Adaptadora de Señalización NOD1/genética , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Receptor Toll-Like 5/genética
3.
Viruses ; 14(11)2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36366583

RESUMEN

The continued evolution of influenza viruses reduces the effectiveness of vaccination and antiviral drugs. The identification of novel and universal agents for influenza prophylaxis and treatment is an urgent need. We have previously described two potent single-domain antibodies (VHH), G2.3 and H1.2, which bind to the stem domain of hemagglutinin and efficiently neutralize H1N1 and H5N2 influenza viruses in vivo. In this study, we modified these VHHs with Fc-fragment to enhance their antiviral activity. Reformatting of G2.3 into bivalent Fc-fusion molecule increased its in vitro neutralizing activity against H1N1 and H2N3 viruses up to 80-fold and, moreover, resulted in obtaining the ability to neutralize H5N2 and H9N2 subtypes. We demonstrated that a dose as low as 0.6 mg/kg of G2.3-Fc or H1.2-Fc administered systemically or locally before infection could protect mice from lethal challenges with both H1N1 and H5N2 viruses. Furthermore, G2.3-Fc reduced the lung viral load to an undetectable level. Both VHH-Fc antibodies showed in vivo therapeutic efficacy when delivered via systemic or local route. The findings support G2.3-Fc as a potential therapeutic agent for both prophylaxis and therapy of Group 1 influenza A infection.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N2 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Anticuerpos de Dominio Único , Ratones , Animales , Humanos , Gripe Humana/prevención & control , Hemaglutininas , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Antivirales/uso terapéutico , Glicoproteínas Hemaglutininas del Virus de la Influenza
4.
Pharmaceutics ; 13(1)2021 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-33406760

RESUMEN

Resistance to antibacterial therapy requires the discovery of new methods for the treatment of infectious diseases. Lactoferrin (LTF) is a well-known naïve first-line defense protein. In the present study, we suggested the use of an adenoviral vector (Ad5) carrying the human gene encoding LTF for direct and cell-mediated gene therapy of maxillofacial area phlegmon in rats. Abscesses were developed by injection of the purulent peritoneal exudate in the molar region of the medial surface of the mandible. At 3-4 days after phlegmon maturation, all rats received ceftriaxone and afterward were subcutaneously injected around the phlegmon with: (1) Ad5 carrying reporter gfp gene encoding green fluorescent protein (Ad5-GFP control group), (2) Ad5 carrying LTF gene (Ad5-LTF group), (3) human umbilical cord blood mononuclear cells (UCBC) transduced with Ad5-GFP (UCBC + Ad5-GFP group), and (4) UCBC transduced with Ad5-LTF (UCBC + Ad5-LTF group). Control rats developed symptoms considered to be related to systemic inflammation and were euthanized at 4-5 days from the beginning of the treatment. Rats from therapeutic groups demonstrated wound healing and recovery from the fifth to seventh day based on the type of therapy. Histological investigation of cervical lymph nodes revealed purulent lymphadenitis in control rats and activated lymphatic tissue in rats from the UCBC + Ad5-LTF group. Our results propose that both approaches of LTF gene delivery are efficient for maxillofacial area phlegmon recovery in rats. However, earlier wound healing and better outcomes in cervical lymph node remodeling in the UCBC + Ad5-LTF group, as well as the lack of direct exposure of the viral vector to the organism, which may cause toxic and immunogenic effects, suggest the benefit of cell-mediated gene therapy.

5.
Protein Expr Purif ; 65(1): 100-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19174190

RESUMEN

The human iron-binding protein lactoferrin (hLf) has been implicated in a number of important physiological pathways, including those regulating immune function and tumor growth. In an effort to develop an efficient system for production of recombinant hLf (rhLf) that is structurally and functionally equivalent to the natural protein, we generated a recombinant CELO (chicken embryo lethal orphan) avian adenovirus containing an expression cassette for hLf. Embryonated chicken eggs were infected with the generated CELO-Lf virus. rhLf expression was measured in the allantoic fluid of infected eggs by ELISA three days later. The level of recombinant protein was about 0.8mg per embryo. rhLf was efficiently purified (up to 85% yield) from the allantoic fluid of infected eggs using affinity chromatography. rhLf produced in the allantoic fluid was characterized in comparison with natural hLf (nhLf) purified from human breast milk. SDS-PAGE, Western blotting and glycosylation analyzes showed that the recombinant protein had similar physical characteristics to nhLf. In addition, we demonstrated that the antioxidative and antimicrobial activity of rhLf produced in this system is equivalent to that of nhLf. Taken together, these results illustrate the utility of the described "recombinant CELO adenovirus-chicken embryo" system for production of functionally active rhLf. Efficient production of rhLf with accurate structure and function is an important step in furthering investigation of Lf as a potential human drug.


Asunto(s)
Aviadenovirus , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/química , Expresión Génica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Alantoides , Animales , Proteínas Portadoras/aislamiento & purificación , Embrión de Pollo , Humanos , Lactoferrina , Proteínas Recombinantes/aislamiento & purificación , Cigoto
6.
PLoS One ; 13(1): e0191574, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29377916

RESUMEN

To avoid outbreaks of influenza virus epidemics and pandemics among human populations, modern medicine requires the development of new universal vaccines that are able to provide protection from a wide range of influenza A virus strains. In the course of development of a universal vaccine, it is necessary to consider that immunity must be generated even against viruses from different hosts because new human epidemic virus strains have their origins in viruses of birds and other animals. We have enriched conserved viral proteins-nucleoprotein (NP) and matrix protein 2 (M2)-by B and T-cell epitopes not only human origin but also swine and avian origin. For this purpose, we analyzed M2 and NP sequences with respect to changes in the sequences of known T and B-cell epitopes and chose conserved and evolutionarily significant epitopes. Eventually, we found consensus sequences of M2 and NP that have the maximum quantity of epitopes that are 100% coincident with them. Consensus epitope-enriched amino acid sequences of M2 and NP proteins were included in a recombinant adenoviral vector. Immunization with Ad5-tet-M2NP induced strong CD8 and CD4 T cells responses, specific to each of the encoded antigens, i.e. M2 and NP. Eight months after immunization with Ad5-tet-M2NP, high numbers of M2- and NP-responding "effector memory" CD44posCD62neg T cells were found in the mouse spleens, which revealed a long-term T cell immune memory conferred by the immunization. In all, the challenge experiments showed an extraordinarily wide-ranging efficacy of protection by the Ad5-tet-M2NP vaccine, covering 5 different heterosubtypes of influenza A virus (2 human, 2 avian and 1 swine).


Asunto(s)
Linfocitos B/inmunología , Epítopos/inmunología , Virus de la Influenza A/inmunología , Nucleoproteínas/inmunología , Linfocitos T/inmunología , Vacunación , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Antivirales/biosíntesis
7.
Front Pharmacol ; 9: 111, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29497380

RESUMEN

Natural brain repair after stroke is extremely limited, and current therapeutic options are even more scarce with no clinical break-through in sight. Despite restricted regeneration in the central nervous system, we have previously proved that human umbilical cord blood mono-nuclear cells (UCB-MC) transduced with adenoviral vectors carrying genes encoding vascular endothelial growth factor (VEGF), glial cell-derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) successfully rescued neurons in amyotrophic lateral sclerosis and spinal cord injury. This proof-of-principle project was aimed at evaluating the beneficial effects of the same triple-gene approach in stroke. Rats subjected to distal occlusion of the middle cerebral artery were treated intrathecally with a combination of these genes either directly or using our cell-based (UCB-MC) approach. Various techniques and markers were employed to evaluate brain injury and subsequent recovery after treatment. Brain repair was most prominent when therapeutic genes were delivered via adenoviral vector- or UCB-MC-mediated approach. Remodeling of brain cortex in the stroke area was confirmed by reduction of infarct volume and attenuated neural cell death, depletion of astrocytes and microglial cells, and increase in the number of oligodendroglial cells and synaptic proteins expression. These results imply that intrathecal injection of genetically engineered UCB-MC over-expressing therapeutic molecules (VEGF, GDNF, and NCAM) following cerebral blood vessel occlusion might represent a novel avenue for future research into treating stroke.

8.
Biomed Res Int ; 2017: 3865802, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28459057

RESUMEN

Chlamydia trachomatis imposes serious health problems and causes infertility. Because of asymptomatic onset, it often escapes antibiotic treatment. Therefore, vaccines offer a better option for the prevention of unwanted inflammatory sequelae. The existence of serologically distinct serovars of C. trachomatis suggests that a vaccine will need to provide protection against multiple serovars. Chlamydia spp. use a highly conserved type III secretion system (T3SS) composed of structural and effector proteins which is an essential virulence factor. In this study, we expressed the T3SS needle protein of Chlamydia muridarum, TC_0037, an ortholog of C. trachomatis CdsF, in a replication-defective adenoviral vector (AdTC_0037) and evaluated its protective efficacy in an intravaginal Chlamydia muridarum model. For better immune responses, we employed a heterologous prime-boost immunization protocol in which mice were intranasally primed with AdTC_0037 and subcutaneously boosted with recombinant TC_0037 and Toll-like receptor 4 agonist monophosphoryl lipid A mixed in a squalene nanoscale emulsion. We found that immunization with TC_0037 antigen induced specific humoral and T cell responses, decreased Chlamydia loads in the genital tract, and abrogated pathology of upper genital organs. Together, our results suggest that TC_0037, a highly conserved chlamydial T3SS protein, is a good candidate for inclusion in a Chlamydia vaccine.


Asunto(s)
Proteínas Bacterianas , Vacunas Bacterianas , Infecciones por Chlamydia , Chlamydia muridarum , Sistemas de Secreción Tipo III , Administración Intranasal , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/prevención & control , Chlamydia muridarum/genética , Chlamydia muridarum/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Enfermedades Vaginales/inmunología , Enfermedades Vaginales/microbiología , Enfermedades Vaginales/prevención & control
9.
Neurosci Lett ; 644: 67-75, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28213069

RESUMEN

Currently, in clinical practice there is no efficient way to overcome the sequences of neurodegeneration after spinal cord traumatic injury. Using a new experimental model of spinal cord contusion injury on miniature pigs, we proposed to deliver therapeutic genes encoding vascular endothelial growth factor (VEGF), glial cell line-derived neurotrophic factor (GDNF) and neural cell adhesion molecule (NCAM) to the damaged area, using umbilical cord blood mononuclear cells (UCBC). In this study, genetically engineered UCBC (2×106 cells in 200 ml of saline) were injected intrathecally to mini-pigs 10days after SCI. Control and experimental mini pigs were observed for 60days after surgery. Histological, electrophysiological, and clinical evaluation demonstrated significant improvement in animal treated with genetically engineered UCBCs. Difference in recovery of the somatosensory evoked potentials and in histological findings in control and treated animals support the positive effect of the gene-cell constriction for recovery after spinal cord injury. Results of this study suggest that transplantation of UCBCs simultaneously transduced with three recombinant adenoviruses Ad5-VEGF, Ad5-GDNF and Ad5-NCAM represent a novel potentially successful approach for treatment of spinal cord injury.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Modelos Animales de Enfermedad , Terapia Genética/métodos , Leucocitos Mononucleares/trasplante , Traumatismos de la Médula Espinal , Adenoviridae/genética , Animales , Femenino , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Moléculas de Adhesión de Célula Nerviosa/genética , Proyectos Piloto , Recuperación de la Función , Porcinos , Porcinos Enanos , Factor A de Crecimiento Endotelial Vascular/genética
10.
Brain Res Bull ; 132: 44-52, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28529158

RESUMEN

Current treatment options for spinal cord injury (SCI) are scarce. One of the most promising innovative approaches include gene-therapy, however no single gene has so far been shown to be of clinical relevance. This study investigates the efficacy of various combinations of vascular endothelial growth factor (VEGF), glial cell-derived neurotrophic factor (GDNF), angiogenin (ANG) and neuronal cell adhesion molecule (NCAM) in rats. Multiple therapeutic genes were administered intrathecally either via adenoviral vectors or by using genetically modified human umbilical cord blood mononuclear cells (hUCBMCs). Following the induction of SCI, serial assessment of cord regeneration was performed, including morphometric analysis of gray and white matters, electrophysiology and behavioral test. The therapeutic gene combinations VEGF+GDNF+NCAM and VEGF+ANG+NCAM had positive outcomes on spinal cord regeneration, with enhanced recovery seen by the cell-based approach when compared to direct gene therapy. The efficacy of the genes and the delivery methods are discussed in this paper, recommending their potential use in SCI.


Asunto(s)
Antígeno CD56/genética , Terapia Genética/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Ribonucleasa Pancreática/genética , Traumatismos de la Médula Espinal/terapia , Factor A de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Antígeno CD56/metabolismo , Trasplante de Células Madre de Sangre del Cordón Umbilical , Modelos Animales de Enfermedad , Escherichia coli , Femenino , Sangre Fetal/citología , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Inyecciones Espinales , Ratas Wistar , Ribonucleasa Pancreática/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Regeneración de la Medula Espinal/fisiología , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
PLoS One ; 11(5): e0155650, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27187797

RESUMEN

Binding of pattern recognition receptors (PRRs) by pathogen-associated molecular patterns (PAMPs) activates innate immune responses and contributes to development of adaptive immunity. Simultaneous stimulation of different types of PRRs can have synergistic immunostimulatory effects resulting in enhanced production of molecules that mediate innate immunity such as inflammatory cytokines, antimicrobial peptides, etc. Here, we evaluated the impact of combined stimulation of PRRs from different families on adaptive immunity by generating alum-based vaccine formulations with ovalbumin as a model antigen and the Toll-like receptor 4 (TLR4) agonist MPLA and the Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) agonist MDP adsorbed individually or together on the alum-ovalbumin particles. Multiple in vitro and in vivo readouts of immune system activation all showed that while individual PRR agonists increased the immunogenicity of vaccines compared to alum alone, the combination of both PRR agonists was significantly more effective. Combined stimulation of TLR4 and NOD2 results in a stronger and broader transcriptional response in THP-1 cells compared to individual PRR stimulation. Immunostimulatory composition containing both PRR agonists (MPLA and MDP) in the context of the alum-based ovalbumin vaccine also enhanced uptake of vaccine particles by bone marrow derived dendritic cells (BMDCs) and promoted maturation (up-regulation of expression of CD80, CD86, MHCII) and activation (production of cytokines) of BMDCs. Finally, immunization of mice with vaccine particles containing both PRR agonists resulted in enhanced cellular immunity as indicated by increased proliferation and activation (IFN-γ production) of splenic CD4+ and CD8+ T cells following in vitro restimulation with ovalbumin and enhanced humoral immunity as indicated by higher titers of ovalbumin-specific IgG antibodies. These results indicate that combined stimulation of TLR4 and NOD2 receptors dramatically enhances activation of both the humoral and cellular branches of adaptive immunity and suggests that inclusion of agonists of these receptors in standard alum-based adjuvants could be used to improve the effectiveness of vaccination.


Asunto(s)
Inmunidad Adaptativa , Adyuvantes Inmunológicos , Inmunogenicidad Vacunal , Proteína Adaptadora de Señalización NOD2/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Receptor Toll-Like 4/inmunología , Acetilmuramil-Alanil-Isoglutamina/inmunología , Línea Celular , Humanos , Inmunidad Celular , Inmunidad Humoral , Lípido A/análogos & derivados , Lípido A/inmunología , Ovalbúmina/inmunología , Receptores de Reconocimiento de Patrones/agonistas , Células TH1/inmunología
12.
PLoS One ; 11(3): e0150958, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26962869

RESUMEN

Developing pathogen-specific recombinant antibody fragments (especially nanobodies) is a very promising strategy for the treatment of infectious disease. Nanobodies have great potential for gene therapy application due to their single-gene nature. Historically, Mycoplasma hominis has not been considered pathogenic bacteria due to the lack of acute infection and partially due to multiple studies demonstrating high frequency of isolation of M. hominis samples from asymptomatic patients. However, recent studies on the role of latent M. hominis infection in oncologic transformation, especially prostate cancer, and reports that M. hominis infects Trichomonas and confers antibiotic resistance to Trichomonas, have generated new interest in this field. In the present study we have generated specific nanobody against M. hominis (aMh), for which the identified target is the ABC-transporter substrate-binding protein. aMh exhibits specific antibacterial action against M. hominis. In an attempt to improve the therapeutic properties, we have developed the adenoviral vector-based gene therapy approach for passive immunization with nanobodies against M. hominis. For better penetration into the mucous layer of the genital tract, we fused aMh with the Fc-fragment of IgG. Application of this comprehensive approach with a single systemic administration of recombinant adenovirus expressing aMh-Fc demonstrated both prophylactic and therapeutic effects in a mouse model of genital M. hominis infection.


Asunto(s)
Adenoviridae , Anticuerpos Antibacterianos/inmunología , Vectores Genéticos , Inmunización Pasiva , Fragmentos Fc de Inmunoglobulinas/inmunología , Infecciones por Mycoplasma/inmunología , Mycoplasma hominis/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Antibacterianos/genética , Camelus/genética , Camelus/inmunología , Femenino , Fragmentos Fc de Inmunoglobulinas/genética , Masculino , Ratones , Ratones Endogámicos DBA , Infecciones por Mycoplasma/prevención & control , Anticuerpos de Dominio Único/genética
13.
Antiviral Res ; 97(3): 245-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23274623

RESUMEN

This work continues a series of recently published studies that employ recombinant single-domain antibody (sdAb, or nanobody®) generation technologies to battle viruses by a passive immunization approach. As a proof of principle, we describe a modified technique to efficiently generate protective molecules against a particular strain of influenza virus within a reasonably short period of time. This approach starts with the immunization of a camel (Camelus bactrianus) with the specified antigen-enriched material presented in as natural a form as possible. An avian influenza virus A/Mallard/Pennsylvania/10218/84 (H5N2) adapted for mice was used as a model source of antigens for both the immunization and phage display-based selection procedures. To significantly increase activities of initially selected monovalent single-domain antibodies, we propose a new type of sdAb formatting that involves the addition of a special type of coiled-coil sequence, the isoleucine zipper domain (ILZ). Presumably, the ILZ-containing peptides adopt trimeric parallel conformations. After the formatting, the biological activities (virus neutralization) of the initially selected anti-influenza virus (H5N2) sdAbs were significantly increased. Intraperitoneal or intranasal administration of the formatted sdAb at 2h before or 24h after viral challenge specifically protects mice from lethal infection with influenza virus. We hope that the described approach combined with the selection focused on particular conservative epitopes will lead to the generation of sdAb-based molecules protective against a broad spectrum of influenza virus subtypes.


Asunto(s)
Anticuerpos Antivirales/inmunología , Técnicas Inmunológicas/métodos , Subtipo H5N2 del Virus de la Influenza A/fisiología , Gripe Humana/prevención & control , Anticuerpos de Dominio Único/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/química , Anticuerpos Antivirales/genética , Camelus/genética , Camelus/inmunología , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H5N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N2 del Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética
14.
Antiviral Res ; 97(3): 318-28, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23274786

RESUMEN

One effective method for the prevention and treatment of influenza infection is passive immunization. In our study, we examined the feasibility of creating an antibody-based preparation with a prolonged protective effect against influenza virus. Single-domain antibodies (sdAbs) specific for influenza virus hemagglutinin were generated. Experiments in mouse models showed 100% survivability for both intranasal sdAbs administration 24h prior to influenza challenge and 24h after infection. sdAb-gene delivery by an adenoviral vector led to gene expression for up to 14days. Protection by a recombinant adenovirus containing the sdAb gene was observed in cases of administration prior to influenza infection (14d-24h). We also demonstrated that the single administration of a combined preparation containing sdAb DNA and protein expanded the protection time window from 14d prior to 48h after influenza infection. This approach and the application of a broad-spectrum sdAbs will allow the development of efficient drugs for the prevention and treatment of viral infections produced by pandemic virus variants and other infections.


Asunto(s)
Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N2 del Virus de la Influenza A/inmunología , Gripe Humana/prevención & control , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Línea Celular , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Inmunización Pasiva , Subtipo H5N2 del Virus de la Influenza A/genética , Gripe Humana/inmunología , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C
15.
J Mol Med (Berl) ; 89(4): 331-41, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21104066

RESUMEN

The recent pandemic threat of the influenza virus makes the increased safety and efficiency of vaccination against the pathogen a most important issue. It has been well established that for maximum protective effect, the vaccination should mimic natural infection. Therefore, recent efforts to develop a new influenza vaccine have focused on intranasal immunization strategies. Intranasal immunization is capable of inducing secretory IgA and serum IgG responses to provide a double defense against mucosal pathogens. On the other hand, it is desirable that a live pathogen is not present in the vaccine. In addition, for optimal induction of the immune responses via the nasal route, efficient and safe mucosal adjuvants are also required. This is possible to attain using an adenoviral vector for vaccine development. Adenoviral vectors are capable of delivering and protecting the antigen encoding sequence. They also possess a natural mechanism for penetrating into the nasal mucous membrane and are capable of activating the innate immune response. This review describes the basic prerequisites for the involvement of recombinant adenoviruses for mucosal (nasal) vaccine development against the influenza virus.


Asunto(s)
Adenoviridae , Vectores Genéticos , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Membrana Mucosa/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Gripe Humana/prevención & control , Sistema Respiratorio/inmunología , Vacunas Sintéticas/inmunología
16.
Oncotarget ; 2(4): 289-97, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21471611

RESUMEN

The origin of chronic inflammation preceding the development of prostate cancer (PCa) remains unknown. We investigated possible involvement of mycoplasma infection in PCa by screening prostate biopsies from two groups of Russian men undergoing PCa diagnosis. M. hominis was detected by standard PCR in 15% of the 125 patients in the first group and by quantitative real-time PCR in 37.4% of the 123 men in the second group. In both groups, stratification of patients according to diagnosis showed that M. hominis was present at three times higher frequency in patients with PCa than in those with benign prostatic hyperplasia. No M. hominis was detected in the prostates of 27 men without detectable prostate disease. In addition, PCa-positive men had higher titers of antibodies against M. hominis and average PSA levels were higher in M. hominis-positive men. These data, together with previous observations linking mycoplasma infection with cell transformation, genomic instability and resistance to apoptosis, suggest that M. hominis infection may be involved in PCa development and may, therefore, be a potential PCa marker and/or target for improved prevention and treatment of this disease.


Asunto(s)
Carcinoma/etiología , Infecciones por Mycoplasma/complicaciones , Mycoplasma hominis/fisiología , Neoplasias de la Próstata/etiología , Anciano , Anciano de 80 o más Años , Anticuerpos/sangre , Anticuerpos/aislamiento & purificación , Biopsia , Carcinoma/epidemiología , Carcinoma/microbiología , Carcinoma/patología , Estudios de Cohortes , Humanos , Masculino , Persona de Mediana Edad , Infecciones por Mycoplasma/epidemiología , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/patología , Mycoplasma hominis/inmunología , Mycoplasma hominis/aislamiento & purificación , Hiperplasia Prostática/diagnóstico , Hiperplasia Prostática/patología , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/microbiología , Neoplasias de la Próstata/patología , Federación de Rusia/epidemiología
17.
J Virol ; 81(18): 9641-52, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17596306

RESUMEN

Vectors based on the chicken embryo lethal orphan (CELO) avian adenovirus (Ad) have two attractive properties for gene transfer applications: resistance to preformed immune responses to human Ads and the ability to grow in chicken embryos, allowing low-cost production of recombinant viruses. However, a major limitation of this technology is that CELO vectors demonstrate decreased efficiency of gene transfer into cells expressing low levels of the coxsackie-Ad receptor (CAR). In order to improve the efficacy of gene transfer into CAR-deficient cells, we modified viral tropism via genetic alteration of the CELO fiber 1 protein. The alphav integrin-binding motif (RGD) was incorporated at two different sites of the fiber 1 knob domain, within an HI-like loop that we identified and at the C terminus. Recombinant fiber-modified CELO viruses were constructed containing secreted alkaline phosphatase (SEAP) and enhanced green fluorescent protein genes as reporter genes. Our data show that insertion of the RGD motif within the HI-like loop of the fiber resulted in significant enhancement of gene transfer into CAR-negative and CAR-deficient cells. In contrast, CELO vectors containing the RGD motif at the fiber 1 C terminus showed reduced transduction of all cell lines. CELO viruses modified with RGD at the HI-like loop transduced the SEAP reporter gene into rabbit mammary gland cells in vivo with an efficiency significantly greater than that of unmodified CELO vector and similar to that of Ad type 5 vector. These results illustrate the potential for efficient CELO-mediated gene transfer into a broad range of cell types through modification of the identified HI-like loop of the fiber 1 protein.


Asunto(s)
Adenovirus A Aviar/genética , Transducción Genética , Proteínas Virales/genética , Secuencias de Aminoácidos/genética , Animales , Células CHO , Embrión de Pollo , Pollos , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Cricetinae , Cricetulus , Adenovirus A Aviar/metabolismo , Genes Reporteros/genética , Terapia Genética , Humanos , Estructura Terciaria de Proteína/genética , Conejos , Receptores Virales/deficiencia , Receptores Virales/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA