Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Virol ; 95(12): e29309, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38100632

RESUMEN

The E6 and E7 proteins of specific subtypes of human papillomavirus (HPV), including HPV 16 and 18, are highly associated with cervical cancer as they modulate cell cycle regulation. The aim of this study was to investigate the potential antitumor effects of a messenger RNA-HPV therapeutic vaccine (mHTV) containing nononcogenic E6 and E7 proteins. To achieve this, C57BL/6j mice were injected with the vaccine via both intramuscular and subcutaneous routes, and the resulting effects were evaluated. mHTV immunization markedly induced robust T cell-mediated immune responses and significantly suppressed tumor growth in both subcutaneous and orthotopic tumor-implanted mouse model, with a significant infiltration of immune cells into tumor tissues. Tumor retransplantation at day 62 postprimary vaccination completely halted progression in all mHTV-treated mice. Furthermore, tumor expansion was significantly reduced upon TC-1 transplantation 160 days after the last immunization. Immunization of rhesus monkeys with mHTV elicited promising immune responses. The immunogenicity of mHTV in nonhuman primates provides strong evidence for clinical application against HPV-related cancers in humans. All data suggest that mHTV can be used as both a therapeutic and prophylactic vaccine.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Vacunas contra Papillomavirus , Neoplasias del Cuello Uterino , Humanos , Femenino , Animales , Ratones , Virus del Papiloma Humano , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/prevención & control , ARN Mensajero/genética , Proteínas E7 de Papillomavirus/genética , Ratones Endogámicos C57BL , Vacunación/métodos , Inmunización , Neoplasias del Cuello Uterino/prevención & control
2.
PLoS Pathog ; 15(8): e1007767, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31437245

RESUMEN

The tad operons encode the machinery required for adhesive Flp (fimbrial low-molecular-weight protein) pili biogenesis. Vibrio vulnificus, an opportunistic pathogen, harbors three distinct tad loci. Among them, only tad1 locus was highly upregulated in in vivo growing bacteria compared to in vitro culture condition. To understand the pathogenic roles of the three tad loci during infection, we constructed single, double and triple tad loci deletion mutants. Interestingly, only the Δtad123 triple mutant cells exhibited significantly decreased lethality in mice. Ultrastructural observations revealed short, thin filamentous projections disappeared on the Δtad123 mutant cells. Since the pilin was paradoxically non-immunogenic, a V5 tag was fused to Flp to visualize the pilin protein by using immunogold EM and immunofluorescence microscopy. The Δtad123 mutant cells showed attenuated host cell adhesion, decreased biofilm formation, delayed RtxA1 exotoxin secretion and subsequently impaired translocation across the intestinal epithelium compared to wild type, which could be partially complemented with each wild type operon. The Δtad123 mutant was susceptible to complement-mediated bacteriolysis, predominantly via the alternative pathway, suggesting stealth hiding role of the Tad pili. Complement depletion by treating with anti-C5 antibody rescued the viable count of Δtad123 in infected mouse bloodstream to the level comparable to wild type strain. Taken together, all three tad loci cooperate to confer successful invasion of V. vulnificus into deeper tissue and evasion from host defense mechanisms, ultimately resulting in septicemia.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Activación de Complemento/inmunología , Fimbrias Bacterianas/fisiología , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Virulencia , Animales , Adhesión Bacteriana , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Ratones , Ratones Endogámicos ICR , Operón , Ratas , Ratas Sprague-Dawley , Vibriosis/genética , Vibriosis/inmunología , Vibriosis/patología , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo
3.
J Allergy Clin Immunol ; 137(2): 426-35, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26303344

RESUMEN

BACKGROUND: Although the hygiene hypothesis suggests that microbial infections could subvert asthma and thus a microbial product might serve as a therapeutic adjuvant for asthma, the relationship between bacterial components and asthma is complex. Recently, low levels of flagellin, the Toll-like receptor (TLR) 5 ligand, have been reported to promote asthma. OBJECTIVE: We show that a therapeutic dose of flagellin suppresses asthma and that the effect occurs through generating regulatory dendritic cells (rDCs) and regulatory T (Treg) cells. METHODS: Ovalbumin (OVA)-induced wild-type and TLR5 knockout asthmatic mice were treated intranasally with a mixture of OVA and 10 µg of a flagellin B (FlaB; of Vibrio vulnificus). OVA/FlaB-treated rDCs were adoptively transferred to mice with OVA-induced asthma. Anti-CD25 mAb was used to deplete Treg cells. A mixture of house dust mite (HDM) and FlaB was used to treat mice with HDM-induced asthma. Blood CD14(+) monocyte-derived dendritic cells from HDM-sensitive asthmatic patients were treated with FlaB and incubated with autologous CD4(+) T cells. RESULTS: An OVA/FlaB mixture ameliorated OVA-induced asthma by inhibiting TH1/TH2/TH17 responses in a TLR5-dependent manner through generating rDCs and Treg cells. The adoptive transfer of OVA/FlaB-treated dendritic cells inhibited OVA-induced asthma, whereas the depletion of CD25(+) cells eliminated the inhibitory effect. A similar effect of FlaB was observed in mice with HDM-induced asthma. In patients with HDM-sensitive asthma, FlaB-treated rDCs inhibited HDM-stimulated TH1/TH2 responses while enhancing Treg cells in an IL-10-dependent manner. CONCLUSION: These findings collectively suggest that flagellin could be used as a tolerogenic adjuvant to treat allergic asthma.


Asunto(s)
Asma/inmunología , Asma/metabolismo , Células Dendríticas/inmunología , Flagelina/inmunología , Inmunomodulación , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Alérgenos/inmunología , Animales , Asma/genética , Asma/patología , Asma/terapia , Estudios de Casos y Controles , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Ligandos , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Pyroglyphidae/inmunología , Linfocitos T Reguladores/metabolismo , Receptor Toll-Like 5/genética , Receptor Toll-Like 5/metabolismo
4.
Infect Immun ; 84(1): 254-65, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26527216

RESUMEN

TonB systems actively transport iron-bound substrates across the outer membranes of Gram-negative bacteria. Vibrio vulnificus CMCP6, which causes fatal septicemia and necrotizing wound infections, possesses three active TonB systems. It is not known why V. vulnificus CMCP6 has maintained three TonB systems throughout its evolution. The TonB1 and TonB2 systems are relatively well characterized, while the pathophysiological function of the TonB3 system is still elusive. A reverse transcription-PCR (RT-PCR) study showed that the tonB1 and tonB2 genes are preferentially induced in vivo, whereas tonB3 is persistently transcribed, albeit at low expression levels, under both in vitro and in vivo conditions. The goal of the present study was to elucidate the raison d'être of these three TonB systems. In contrast to previous studies, we constructed in-frame single-, double-, and triple-deletion mutants of the entire structural genes in TonB loci, and the changes in various virulence-related phenotypes were evaluated. Surprisingly, only the tonB123 mutant exhibited a significant delay in killing eukaryotic cells, which was complemented in trans with any TonB operon. Very interestingly, we discovered that flagellum biogenesis was defective in the tonB123 mutant. The loss of flagellation contributed to severe defects in motility and adhesion of the mutant. Because of the difficulty of making contact with host cells, the mutant manifested defective RtxA1 toxin production, which resulted in impaired invasiveness, delayed cytotoxicity, and decreased lethality for mice. Taken together, these results indicate that a series of virulence defects in all three TonB systems of V. vulnificus CMCP6 coordinately complement each other for iron assimilation and full virulence expression by ensuring flagellar biogenesis.


Asunto(s)
Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Proteínas de la Membrana/genética , Vibrio vulnificus/patogenicidad , Animales , Proteínas Bacterianas/biosíntesis , Toxinas Bacterianas/biosíntesis , Transporte Biológico/genética , Línea Celular Tumoral , Femenino , Flagelos/genética , Células HeLa , Humanos , Hierro/metabolismo , Proteínas de la Membrana/biosíntesis , Ratones , Cavidad Peritoneal/microbiología , Ratas , Ratas Sprague-Dawley , Vibriosis/microbiología , Vibrio vulnificus/genética , Vibrio vulnificus/crecimiento & desarrollo
5.
J Transl Med ; 14(1): 135, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27184355

RESUMEN

BACKGROUND: Noroviruses (NoVs) are a major cause of childhood gastroenteritis and foodborne diseases worldwide. Lack of appropriate animal models or cell-based culture systems makes the development and evaluation of NoV-specific vaccines a daunting task. VP1 is the major capsid protein of the NoVs that acts as a binding motif to human histo-blood group antigens (HBGAs) through its protruding 2 (P2) domain and can serve as a protective antigen candidate for vaccine development. METHODS: Recombinantly produced NoV specific P domain (Pd) vaccine was inoculated into groups of mice either alone or in conjugation with mucosal adjuvant FlaB, the flagellar protein from Vibrio vulnificus. Antigen specific humoral and cell mediated immune responses were assessed by enzyme linked immunosorbent assay (ELISA) or fluorescent activated cell sorting (FACS). A comparative analysis of various routes of vaccination viz. intranasal, sublingual and subcutaneous, was also done. RESULTS: In this study, we show that a recombinant Pd-vaccine administered through intranasal route induced a robust TH2-dependent humoral immune response and that the combination of vaccine with FlaB significantly enhanced the antibody response. Interestingly, FlaB induced a mixed TH1/TH2 type of immune response with a significant induction of IgG1 as well as IgG2a antibodies. FlaB also induced strong IgA responses in serum and feces. FlaB mediated antibody responses were toll like receptor 5 (TLR5) dependent, since the FlaB adjuvanticity was lost in TLR5(-/-) mice. Further, though the Pd-vaccine by itself failed to induce a cell mediated immune response, the Pd-FlaB combination stimulated a robust CD4(+)IFNγ(+) and CD8(+)IFNγ(+) T cell response in spleen and mesenteric lymph nodes. We also compared the adjuvant effects of FlaB with that of alum and complete Freund's adjuvant (CFA). We found that subcutaneously inoculated FlaB induced more significant levels of IgG and IgA in both serum and feces compared to alum or CFA in respective samples. CONCLUSION: We validate the use of TLR5 agonist as a strong mucosal adjuvant that would facilitate the development of NoV specific vaccines for humans and veterinary use. This study also highlights the importance of route of immunization in inducing the appropriate immune responses in mucosal compartments.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Flagelina/farmacología , Inmunidad Mucosa , Norovirus/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología , Animales , Antígenos Virales/inmunología , Clonación Molecular , Dimerización , Heces , Femenino , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Ratones Endogámicos BALB C , Norovirus/efectos de los fármacos , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Receptor Toll-Like 5/metabolismo , Vacunación
6.
Infect Immun ; 82(1): 29-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24101693

RESUMEN

Vibrio vulnificus is a halophilic pathogenic bacterium that is motile due to the presence of a single polar flagellum. V. vulnificus possesses a total of six flagellin genes organized into two loci (flaFBA and flaCDE). We proved that all six of the flagellin genes were transcribed, whereas only five (FlaA, -B, -C, -D, and -F) of the six flagellin proteins were detected. To understand roles of the six V. vulnificus flagellins in motility and virulence, mutants with single and multiple flagellin deletions were constructed. Mutations in flaB or flaC or the flaCDE locus resulted in a significant decrease in motility, adhesion, and cytotoxicity, whereas single mutations in the other flagellin genes or the flaFBA locus showed little or no effect. The motility was completely abolished only in the mutant lacking all six flagellin genes (flaFBA flaCDE). Surprisingly, a double mutation of flaB and flaD, a gene sharing 99% identity with the flaB at the amino acid level, resulted in the largest decrease in motility, adhesion, and cytotoxicity except for the mutant in which all six genes were deleted (the hexa mutant). Additionally, the 50% lethal doses (LD50s) of the flaB flaD and the flaFBA flaCDE mutants increased 23- and 91-fold in a mouse model, respectively, and the in vitro and in vivo invasiveness of the mutants was significantly decreased compared to that of the wild type. Taken together, the multiple flagellin subunits differentially contribute to the flagellum biogenesis and the pathogenesis of V. vulnificus, and among the six flagellin genes, flaB, flaD, and flaC were the most influential components.


Asunto(s)
Flagelina/genética , Vibriosis/fisiopatología , Vibrio vulnificus/fisiología , Animales , Adhesión Bacteriana/fisiología , Modelos Animales de Enfermedad , Flagelos/química , Flagelos/genética , Flagelina/metabolismo , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/fisiología , Ratones , Mutación , Vibriosis/genética , Vibrio vulnificus/genética , Vibrio vulnificus/patogenicidad
7.
J Infect Dis ; 207(9): 1406-15, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23225896

RESUMEN

Vibrio vulnificus, a halophilic estuarine bacterium causing fatal septicemia and necrotic wound infection, is highly cytotoxic to eukaryotic cells. We have reported that RtxA1 toxin kills host cells only after they come into contact with bacteria and plays an essential role in the pathogenesis of V. vulnificus. This study was performed to elucidate the mechanism by which the RtxA1 toxin mediates the death of HeLa cells. By using confocal microscopy and immunoblot analysis, we show that the 501-kDa RtxA1 toxin is processed into 2 fragments after its secretion into host cells. The largerN-terminal fragment (RtxA1-N; approximately 370 kDa) remained at the host cell membrane, whereas the smaller C-terminal fragment (RtxA1-C; approximately 130 kDa) was internalized into the host cell cytoplasm. RtxA1-N is believed to polymerize and form pores at the host cell membrane and to induce an increase in necrotic volume related to calcium. The RtxA1 toxin caused an increase in the intracellular Ca(2+) concentration and the subsequent activation of JNK. The cell death mechanism occurred via calcium-dependent mitochondrial pathways, which caused calcium sequestration in the mitochondria, accompanied by irreversible mitochondrial membrane dysfunction and adenosine triphosphate depletion, and was later accompanied by the disruption of the integrity of the plasma membrane.


Asunto(s)
Apoptosis , Toxinas Bacterianas/metabolismo , Calcio/metabolismo , Citotoxinas/metabolismo , Mitocondrias/efectos de los fármacos , Vibrio vulnificus/patogenicidad , Adenosina Trifosfato/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células HeLa , Humanos , Immunoblotting , Microscopía Confocal , Mitocondrias/fisiología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Transporte de Proteínas
8.
Front Immunol ; 15: 1375767, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646546

RESUMEN

Cancer immunotherapy has made tremendous advancements in treating various malignancies. The biggest hurdle to successful immunotherapy would be the immunosuppressive tumor microenvironment (TME) and low immunogenicity of cancer cells. To make immunotherapy successful, the 'cold' TME must be converted to 'hot' immunostimulatory status to activate residual host immune responses. To this end, the immunosuppressive equilibrium in TME should be broken, and immunogenic cancer cell death ought to be induced to stimulate tumor-killing immune cells appropriately. Photodynamic therapy (PDT) is an efficient way of inducing immunogenic cell death (ICD) of cancer cells and disrupting immune-restrictive tumor tissues. PDT would trigger a chain reaction that would make the TME 'hot' and have ICD-induced tumor antigens presented to immune cells. In principle, the strategic combination of PDT and immunotherapy would synergize to enhance therapeutic outcomes in many intractable tumors. Novel technologies employing nanocarriers were developed to deliver photosensitizers and immunotherapeutic to TME efficiently. New-generation nanomedicines have been developed for PDT immunotherapy in recent years, which will accelerate clinical applications.


Asunto(s)
Inmunoterapia , Nanopartículas , Neoplasias , Fotoquimioterapia , Fármacos Fotosensibilizantes , Microambiente Tumoral , Fotoquimioterapia/métodos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Animales , Fármacos Fotosensibilizantes/uso terapéutico , Terapia Combinada , Nanomedicina/métodos
9.
NPJ Vaccines ; 9(1): 108, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879560

RESUMEN

Alzheimer's disease (AD) and related tauopathies are associated with pathological tau protein aggregation, which plays an important role in neurofibrillary degeneration and dementia. Targeted immunotherapy to eliminate pathological tau aggregates is known to improve cognitive deficits in AD animal models. The tau repeat domain (TauRD) plays a pivotal role in tau-microtubule interactions and is critically involved in the aggregation of hyperphosphorylated tau proteins. Because TauRD forms the structural core of tau aggregates, the development of immunotherapies that selectively target TauRD-induced pathological aggregates holds great promise for the modulation of tauopathies. In this study, we generated recombinant TauRD polypeptide that form neurofibrillary tangle-like structures and evaluated TauRD-specific immune responses following intranasal immunization in combination with the mucosal adjuvant FlaB. In BALB/C mice, repeated immunizations at one-week intervals induced robust TauRD-specific antibody responses in a TLR5-dependent manner. Notably, the resulting antiserum recognized only the aggregated form of TauRD, while ignoring monomeric TauRD. The antiserum effectively inhibited TauRD filament formation and promoted the phagocytic degradation of TauRD aggregate fragments by microglia. The antiserum also specifically recognized pathological tau conformers in the human AD brain. Based on these results, we engineered a built-in flagellin-adjuvanted TauRD (FlaB-TauRD) vaccine and tested its efficacy in a P301S transgenic mouse model. Mucosal immunization with FlaB-TauRD improved quality of life, as indicated by the amelioration of memory deficits, and alleviated tauopathy progression. Notably, the survival of the vaccinated mice was dramatically extended. In conclusion, we developed a mucosal vaccine that exclusively targets pathological tau conformers and prevents disease progression.

10.
Nat Commun ; 15(1): 46, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167804

RESUMEN

Addressing age-related immunological defects through therapeutic interventions is essential for healthy aging, as the immune system plays a crucial role in controlling infections, malignancies, and in supporting tissue homeostasis and repair. In our study, we show that stimulating toll-like receptor 5 (TLR5) via mucosal delivery of a flagellin-containing fusion protein effectively extends the lifespan and enhances the healthspan of mice of both sexes. This enhancement in healthspan is evidenced by diminished hair loss and ocular lens opacity, increased bone mineral density, improved stem cell activity, delayed thymic involution, heightened cognitive capacity, and the prevention of pulmonary lung fibrosis. Additionally, this fusion protein boosts intestinal mucosal integrity by augmenting the surface expression of TLR5 in a certain subset of dendritic cells and increasing interleukin-22 (IL-22) secretion. In this work, we present observations that underscore the benefits of TLR5-dependent stimulation in the mucosal compartment, suggesting a viable strategy for enhancing longevity and healthspan.


Asunto(s)
Longevidad , Receptor Toll-Like 5 , Animales , Ratones , Flagelina/metabolismo , Mucosa Intestinal/metabolismo , Longevidad/genética , Pulmón/metabolismo
11.
J Biol Chem ; 287(22): 18182-9, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22496368

RESUMEN

STAT3 was recently reported to suppress tumor invasion in Apc(min)(/+) mice. We investigated the mechanisms by which STAT3 inhibits intestinal epithelial tumors using Apc(min)(/+)/Stat3(IEC-KO) mice (intestinal epithelial cell (IEC)-specific deletion of STAT3 in the Apc(min)(/+) background) to determine the role of STAT3 in carcinogenesis in vivo as well as colorectal cancer cell lines in vitro. To inhibit invasion of IEC tumors, STAT3 functions as a molecular adaptor rather than a transcription factor. Accordingly, the tumors in Apc(min)(/+)/Stat3(IEC-KO) mice undergo adenoma-to-carcinoma transition and acquire an invasive phenotype. Similarly, STAT3 knockdown in a colorectal cell line enhances IEC invasion. We demonstrate that STAT3 down-regulates SNAI (Snail-1) expression levels and hence suppresses epithelial-mesenchymal transition of colorectal cancer cells. Mechanistically, STAT3 facilitates glycogen synthase kinase (GSK) 3ß-mediated degradation of SNAI by regulating phosphorylation of GSK3ß. Our data identified a new role for STAT3 in the adenoma-to-carcinoma sequence of intestinal tumors.


Asunto(s)
Adenoma/patología , Carcinoma/patología , Genes APC , Factor de Transcripción STAT3/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Proliferación Celular , Cartilla de ADN , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Neoplasias Intestinales/patología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Factor de Transcripción STAT3/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
12.
J Biol Chem ; 287(23): 18888-99, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22493443

RESUMEN

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) is an orphan nuclear receptor of the steroid-thyroid hormone receptor superfamily. COUP-TFII is widely expressed in multiple tissues and organs throughout embryonic development and has been shown to regulate cellular growth, differentiation, and organ development. However, the role of COUP-TFII in osteoblast differentiation has not been systematically evaluated. In the present study, COUP-TFII was strongly expressed in multipotential mesenchymal cells, and the endogenous expression level decreased during osteoblast differentiation. Overexpression of COUP-TFII inhibited bone morphogenetic protein 2 (BMP2)-induced osteoblastic gene expression. The results of alkaline phosphatase, Alizarin Red staining, and osteocalcin production assay showed that COUP-TFII overexpression blocks BMP2-induced osteoblast differentiation. In contrast, the down-regulation of COUP-TFII synergistically induced the expression of BMP2-induced osteoblastic genes and osteoblast differentiation. Furthermore, the immunoprecipitation assay showed that COUP-TFII and Runx2 physically interacted and COUP-TFII significantly impaired the Runx2-dependent activation of the osteocalcin promoter. From the ChIP assay, we found that COUP-TFII repressed DNA binding of Runx2 to the osteocalcin gene, whereas Runx2 inhibited COUP-TFII expression via direct binding to the COUP-TFII promoter. Taken together, these findings demonstrate that COUP-TFII negatively regulates osteoblast differentiation via interaction with Runx2, and during the differentiation state, BMP2-induced Runx2 represses COUP-TFII expression and promotes osteoblast differentiation.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Diferenciación Celular/fisiología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Osteoblastos/metabolismo , Animales , Factor de Transcripción COUP II/genética , Línea Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica/fisiología , Humanos , Metaloproteinasa 2 de la Matriz/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoblastos/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Unión Proteica
13.
Gastroenterology ; 143(1): 145-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22475534

RESUMEN

BACKGROUND & AIMS: Foxp3(+) T-regulatory cells (Tregs) maintain intestinal homeostasis under conditions of continuous challenge with inflammatory microbes. However, plasticity of the Treg population under certain conditions has been reported; Foxp3(+) Tregs can be converted to Foxp3(-) CD4(+) T cells. METHODS: We used mice with a T cell-induced colitis model to study the regulatory role of type I interferons (IFNs) in adaptive immunity. We transferred CD4(+)CD45RB(hi) (RB(hi)) T cells, with or without CD4(+)CD45RB(lo) CD25(+) T cells, from wild-type or IFN-αßR(-/-) mice into Rag1(-/-) recipients. We analyzed induction of colitis by flow cytometry, confocal microscopy, and enzyme-linked immunosorbent assay and reverse-transcription polymerase chain reaction analyses. IFN-αßR(-/-)Rag(-/-) mice were given injections of recombinant IFN-α following transfer of IFN-αßR(-/-) RB(hi) T cells and CD4(+)Foxp3(+) cells from Foxp3-eGFP mice. RESULTS: Signaling by type I IFNs was required for maintenance of Foxp3 expression and the suppressive activity of Tregs in mice. Transfer of CD4(+)CD45RB(lo)CD25(+) Tregs from IFN-αßR(-/-) mice did not prevent T-cell induction of colitis in mice. Foxp3 expression by Tregs transferred from IFN-αßR(-/-) mice was significantly lower than that of Tregs from wild-type mice. Administration of recombinant IFN-α reduced T cell-mediated colitis by increasing the number of Foxp3(+) Tregs and their suppressive functions. CONCLUSIONS: Type I IFNs regulate intestinal homeostasis by maintaining Foxp3 expression on Tregs in colons of mice under inflammatory conditions.


Asunto(s)
Colitis/inmunología , Factores de Transcripción Forkhead/biosíntesis , Interferón Tipo I/inmunología , Linfocitos T Reguladores/inmunología , Inmunidad Adaptativa , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL
14.
Microbiol Immunol ; 57(4): 273-80, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23586631

RESUMEN

Vibrio vulnificus causes fatal septicemia in susceptible subjects after the ingestion of raw seafood. In the present study, the roles of cyclic adenosine monophosphate (cAMP) receptor protein (CRP) in V. vulnificus pathogenesis were investigated. A mutation in the V. vulnificus crp gene resulted in a significant down-regulation of various virulence phenotypes, except for RtxA1-mediated cytoskeletal rearrangement. Bacterial growth was impeded by the crp mutation. In addition, colony morphology was converted from opaque to translucent type by this mutation, which implies a decrease in capsule production. The crp mutant also showed significant decrease in motility and adhesion to host cells. V. vulnificus CRP positively regulated production of hemolysin and protease at transcriptional level. All these changes in the crp mutant were fully complemented in trans by a plasmid harboring the wild-type gene. In contrast, CRP negatively regulated the expression of RtxA1. The crp mutant caused the cytoskeletal rearrangement in HeLa cells, which is a hallmark activity of RtxA1 toxin. Taken together, CRP seems to play a dual regulatory role in various virulence traits of V. vulnificus.


Asunto(s)
Proteína Receptora de AMP Cíclico/genética , Regulación Bacteriana de la Expresión Génica , Vibrio vulnificus/fisiología , Vibrio vulnificus/patogenicidad , Virulencia/genética , Animales , Adhesión Bacteriana , Línea Celular , Proteína Receptora de AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Femenino , Células HeLa , Proteínas Hemolisinas/biosíntesis , Humanos , Ratones , Mutación , Péptido Hidrolasas/biosíntesis , Conejos , Vibriosis/microbiología
15.
Curr Opin Virol ; 60: 101330, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37084463

RESUMEN

Flagellin is the cognate ligand for host pattern recognition receptors, toll-like receptor 5 (TLR5) in the cell surface, and NAIP5/NLRC4 inflammasome in the cytosol. TLR5-binding domain is located in D1 domain, where crucial amino acid sequences are conserved among diverse bacteria. The highly conserved C-terminal 35 amino acids of flagellin were proved to be responsible for the inflammasome activation by binding to NAIP5. D2/D3 domains, located in the central region and exposed to the outside surface of flagellar filament, are heterogeneous across bacterial species and highly immunogenic. Taking advantage of TLR5- and NLRC4-stimulating activities, flagellin has been actively developed as a vaccine adjuvant and immunotherapeutic. Because of its immunogenicity, there exist worries concerning diminished efficacy and possible reactogenicity after repeated administration. Deimmunization of flagellin derivatives while preserving the TLR5/NLRC4-mediated immunomodulatory activity should be the most reasonable option for clinical application. This review describes strategies and current achievements in flagellin deimmunization.


Asunto(s)
Inflamasomas , Receptor Toll-Like 5 , Receptor Toll-Like 5/metabolismo , Inmunidad Innata , Flagelina/genética , Flagelina/química , Bacterias/metabolismo
16.
NPJ Vaccines ; 8(1): 139, 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752138

RESUMEN

Flagellin, the TLR5 agonist, shows potent adjuvant activities in diverse vaccines and immunotherapies. Vibrio vulnificus flagellin B expressed in eukaryotic cells (eFlaB) could not stimulate TLR5 signaling. Enzymatic deglycosylation restored eFlaB's TLR5 stimulating functionality, suggesting that glycosylation interferes with eFlaB binding to TLR5. Site-directed mutagenesis of N-glycosylation residues restored TLR5 stimulation and adjuvanticity. Collectively, deglycosylated eFlaB may provide a built-in adjuvant platform for eukaryotic-expressed antigens and nucleic acid vaccines.

17.
Biomaterials ; 286: 121542, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35594837

RESUMEN

Therapeutic cancer vaccines (TCVs) should induce robust tumor-specific T cell responses. To achieve this, TCVs incorporate T cell epitopes and strong adjuvants. Here, we report an all-in-one adjuvanted cancer vaccine platform that targets the intracellular compartment of dentritic cells and subsequently induces effective cytotoxic T cell responses. We screened a novel peptide (DCpep6) that specifically binds and transmits into CD11c+ cells through a novel in vivo phage biopanning. We then engineered a protein-based TCV (DEF) consisting of DCpep6 (D), an optimized HPV E7 tumor antigen (E), and a built-in flagellin adjuvant (F) as a single molecule. DEF was stably expressed, and each component was functional. In vivo-administered DEF rapidly biodistributed in draining LNs and internalized into CD11c+ cells. DEF immunization elicited strong antitumor T cell responses and provided long-term survival of TC-1 tumor-implanted mice. The DEF-mediated antitumor effect was abolished in NLRC4-/- mice. Taken together, we propose a protein-based all-in-one TCV platform that intracellularly codelivers tumor antigen and inflammasome activator to DCs to induce long-lasting antitumor T cell responses.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Adyuvantes Inmunológicos/farmacología , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos , Citosol , Células Dendríticas , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias/metabolismo
18.
Transl Cancer Res ; 11(6): 1595-1602, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35836530

RESUMEN

Background: Therapeutic cancer vaccines, which induce or amplify tumor-specific T cell responses, are a critical component of multiple combination cancer immunotherapy regimens. Innovative neoantigen identification continually prompts the development of vaccine platforms. However, vaccine monotherapy is not sufficient to eradicate tumors. Thus, therapeutic strategies combining cancer vaccines and treatment with other immune modulators have been expl, ored. Previously, we showed that flagellin has an excellent adjuvant activity to induce effective immune responses to co-administered peptide epitopes through TLR5 stimulation in mouse TC-1 tumor models and flagellin-expressing bacteria modulate the tumor microenvironment (TME) toward enhanced immunogenicity. Methods: Given that short- and long-peptides undergo different fates of internalization, processing, and MHC-restricted presentation by professional antigen-presenting cells (APCs), we compared the antitumor activity of flagellin-adjuvanted peptide vaccines by employing the E7 CD8 epitope short peptide (E7-SP49-57) and E7 long peptides (E7-LP2043-62 and E7-LP3543-77). Because combinations take center stage in immune checkpoint inhibitor (ICI) therapy, we evaluated the best E7 peptide vaccine component for combination with anti-PD-1 in the mouse TC-1 model. Results: Flagellin adjuvanted E7-LP35 vaccine (FlaB-LP35Vax) showed significantly higher antitumor activity than flagellin adjuvanted E7-SP vaccine (FlaB-SPVax) and flagellin adjuvanted E7-LP20 vaccine (FlaB-LP20Vax) in a mouse TC-1 tumor model. Coadministration of flagellin was essential for E7-mediated tumor suppression. PD-1 blockade enhanced the therapeutic efficacy of FlaB-LP35Vax but not FlaB-SPVax. Taken together, E7-LP35 is an optimal tumor antigen for flagellin-adjuvanted E7 cancer vaccines, and the combination of FlaB-LP35Vax with anti-PD-1 antibody treatment induced long-term antitumor immune responses. Conclusions: This result suggests that cooperation between CD4+ and CD8+ cell-mediated immune responses is essential for the success of combination therapy with cancer vaccines and ICIs.

19.
J Infect Dis ; 201(1): 97-105, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19919301

RESUMEN

Infection with the human pathogen Vibrio vulnificus leads to the generation of reactive oxygen species (ROS) via NAD(P)H oxidase (Nox) in host cells. In the present study, we employed mutant V. vulnificus strains to identify an essential virulence factor responsible for this ROS generation. We found that repeats-in-toxin A1 (RtxA1) expressed by V. vulnificus acts via Nox1 to induce significant ROS generation in the intestine epithelial cells, which ultimately results in cell death. Furthermore, RtxA1 modulates the small GTPase Rac2, which is known to play an important role in the activation of Nox. When mice were infected by the oral method, in contrast with the wild-type bacteria, an RtxA1-deficient V. vulnificus mutant was unable to induce ROS generation within the intestine and failed to cause death. These findings strongly suggest that RtxA1-induced Rac2 expression is a critical step underlying the pathogenicity of V. vulnificus.


Asunto(s)
Toxinas Bacterianas/metabolismo , Células Epiteliales/microbiología , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Proteínas de Unión al GTP rac/metabolismo , Animales , Células CACO-2 , Humanos , Mucosa Intestinal/microbiología , Ratones , NADPH Oxidasa 1 , NADPH Oxidasas , Especies Reactivas de Oxígeno/metabolismo , Vibrio vulnificus/metabolismo , Proteína RCA2 de Unión a GTP
20.
NPJ Vaccines ; 6(1): 116, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518537

RESUMEN

Flagellin, a protein-based Toll-like receptor agonist, is a versatile adjuvant applicable to wide spectrum of vaccines and immunotherapies. Given reiterated treatments of immunogenic biopharmaceuticals should lead to antibody responses precluding repeated administration, the development of flagellin not inducing specific antibodies would greatly expand the chances of clinical applications. Here we computationally identified immunogenic regions in Vibrio vulnificus flagellin B and deimmunized by simply removing a B cell epitope region. The recombinant deimmunized FlaB (dFlaB) maintains stable TLR5-stimulating activity. Multiple immunization of dFlaB does not induce FlaB-specific B cell responses in mice. Intranasally co-administered dFlaB with influenza vaccine enhanced strong Ag-specific immune responses in both systemic and mucosal compartments devoid of FlaB-specific Ab production. Notably, dFlaB showed better protective immune responses against lethal viral challenge compared with wild type FlaB. The deimmunizing B cell epitope deletion did not compromise stability and adjuvanticity, while suppressing unwanted antibody responses that may negatively affected vaccine antigen-directed immune responses in repeated vaccinations. We explain the underlying mechanism of deimmunization by employing molecular dynamics analysis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA