Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Infect Immun ; 89(1)2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33139380

RESUMO

Chlamydia trachomatis is an obligate intracellular pathogen that causes sexually transmitted disease. In women, chlamydial infections may cause pelvic inflammatory disease (PID), ectopic pregnancy, and infertility. The role of antibodies in protection against a primary Chlamydia infection is unclear and was a focus of this work. Using the C. muridarum mouse infection model, we show that intestinal mucosa is infected via intranasal (i.n.) or per-oral (p.o.) Chlamydia inoculation and that unlike the female reproductive tract (FRT) mucosa, it halts systemic Chlamydia dissemination. Moreover, p.o. immunization or infection with Chlamydia confers protection against per-vaginal (p.v.) challenge, resulting in significantly decreased bacterial burden in the FRT, accelerated Chlamydia clearance, and reduced hydrosalpinx pathology. In contrast, subcutaneous (s.c.) immunization conferred no protection against the p.v. challenge. Both p.o. and s.c. immunizations induced Chlamydia-specific serum IgA. However, IgA was found only in the vaginal washes and fecal extracts of p.o.-immunized animals. Following a p.v. challenge, unimmunized control and s.c.-s.c.-immunized animals developed Chlamydia-specific intestinal IgA yet failed to develop IgA in the FRT, indicating that IgA response in the FRT relies on the FRT to gastrointestinal tract (GIT) antigen transport. Vaginal secretions of p.o.-immunized animals neutralize Chlamydia in vivo, resulting in significantly lower Chlamydia burden in the FRT and Chlamydia transport to the GIT. We also show that infection of the GIT is not necessary for induction of protective immunity in the FRT, a finding that is important for the development of p.o. subunit vaccines to target Chlamydia and possibly other sexually transmitted pathogens.


Assuntos
Infecções por Chlamydia/imunologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/imunologia , Genitália Feminina/imunologia , Genitália Feminina/microbiologia , Imunoglobulina A Secretora/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos/imunologia , Carga Bacteriana , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/prevenção & controle , Feminino , Interações Hospedeiro-Patógeno/imunologia , Imunidade nas Mucosas , Imunização , Camundongos
2.
Biomedicines ; 8(9)2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32962144

RESUMO

Soft 3D-fibrin-gel selected tumor repopulating cells (TRCs) from the B16F1 melanoma cell line exhibit extraordinary self-renewal and tumor-regeneration capabilities. However, their biomarkers and gene regulatory features remain largely unknown. Here, we utilized the next-generation sequencing-based RNA sequencing (RNA-seq) technique to discover novel biomarkers and active gene regulatory features of TRCs. Systems biology analysis of RNA-seq data identified differentially expressed gene clusters, including the cell adhesion cluster, which subsequently identified highly specific and novel biomarkers, such as Col2a1, Ncam1, F11r, and Negr1. We validated the expression of these genes by real-time qPCR. The expression level of Col2a1 was found to be relatively low in TRCs but twenty-fold higher compared to the parental control cell line, thus making the biomarker very specific for TRCs. We validated the COL2A1 protein by immunofluorescence microscopy, showing a higher expression of COL2A1 in TRCs compared to parental control cells. KEGG pathway analysis showed the JAK/STAT, hypoxia, and Akt signaling pathways to be active in TRCs. Besides, the aerobic glycolysis pathway was found to be very active, indicating a typical Warburg Effect on highly tumorigenic cells. Together, our study revealed highly specific biomarkers and active cell signaling pathways of melanoma TRCs that can potentially target and neutralize TRCs.

3.
PLoS Pathog ; 15(12): e1008207, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790512

RESUMO

Chlamydia trachomatis is a Gram-negative bacterial pathogen and a major cause of sexually transmitted disease and preventable blindness. In women, infections with C. trachomatis may lead to pelvic inflammatory disease (PID), ectopic pregnancy, chronic pelvic pain, and infertility. In addition to infecting the female reproductive tract (FRT), Chlamydia spp. are routinely found in the gastro-intestinal (GI) tract of animals and humans and can be a reservoir for reinfection of the FRT. Whether Chlamydia disseminates from the FRT to the GI tract via internal routes remains unknown. Using mouse-specific C. muridarum as a model pathogen we show that Chlamydia disseminates from the FRT to the GI tract in a stepwise manner, by first infecting the FRT-draining iliac lymph nodes (ILNs), then the spleen, then the GI tract. Tissue CD11c+ DCs mediate the first step: FRT to ILN Chlamydia transport, which relies on CCR7:CCL21/CCL19 signaling. The second step, Chlamydia transport from ILN to the spleen, also relies on cell transport. However, this step is dependent on cell migration mediated by sphingosine 1-phosphate (S1P) signaling. Finally, spleen to GI tract Chlamydia spread is the third critical step, and is significantly hindered in splenectomized mice. Inhibition of Chlamydia dissemination significantly reduces or precludes the induction of Chlamydia-specific serum IgG antibodies, presence of which is correlated with FRT pathology in women. This study reveals important insights in context of Chlamydia spp. pathogenesis and will inform the development of therapeutic targets and vaccines to combat this pathogen.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/patogenicidade , Trato Gastrointestinal/microbiologia , Genitália Feminina/microbiologia , Infecções do Sistema Genital/microbiologia , Animais , Feminino , Camundongos
4.
Vaccine ; 36(8): 1126-1131, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29406243

RESUMO

The purpose of this study was to determine if conjugating a recombinant Eimeria maxima protein, namely EmaxIMP1, into 20 nm polystyrene nanoparticles (NP) could improve the level of protective immunity against E. maxima challenge infection. Recombinant EmaxIMP1 was expressed in Escherichia coli as a poly-His fusion protein, purified by NiNTA chromatography, and conjugated to 20 nm polystyrene NP (NP-EmaxIMP1). NP-EMaxIMP1 or control non-recombinant (NP-NR) protein were delivered per os to newly-hatched broiler chicks with subsequent booster immunizations at 3 and 21 days of age. In battery cage studies (n = 4), chickens immunized with NP-EMaxIMP1 displayed complete protection as measured by weight gain (WG) against E. maxima challenge compared to chickens immunized with NP-NR. WG in the NP-EMaxIMP1-immunized groups was identical to WG in chickens that were not infected with E. maxima infected chickens. In floor pen studies (n = 2), chickens immunized with NP-EMaxIMP1 displayed partial protection as measured by WG against E. maxima challenge compared to chickens immunized with NP-NR. In order to understand the basis for immune stimulation, newly-hatched chicks were inoculated per os with NP-EMaxIMP1 or NP-NR protein, and the small intestine, bursa, and spleen, were examined for NP localization at 1 h and 6 h post-inoculation. Within 1 h, both NP-EMaxIMP1 and NP-NR were observed in all 3 tissues. An increase was observed in the level of NP-EmaxIMP1 and NP-NR in all tissues at 6 h post-inoculation. These data indicate that 20 nm NP-EmaxIMP1 or NP-NR reached deeper tissues within hours of oral inoculation and elicited complete to partial immunity against E. maxima challenge infection.


Assuntos
Antígenos de Protozoários/imunologia , Coccidiose/prevenção & controle , Coccidiose/veterinária , Eimeria/imunologia , Nanopartículas/química , Doenças das Aves Domésticas/prevenção & controle , Vacinas Protozoárias/imunologia , Administração Oral , Animais , Antígenos de Protozoários/química , Galinhas , Tamanho da Partícula , Poliestirenos/química , Doenças das Aves Domésticas/parasitologia , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Vacinação
5.
J Biomed Mater Res A ; 105(2): 557-565, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27750375

RESUMO

In this work, we report a protocol for synthesizing nanosize ovalbumin-functionalized polydiacetylene (PDA) liposomes (LP-Ova). We show that LP-Ova administered per-orally (p.o.) and subcutaneously (s.c.), without the use of adjuvants, induces high serum IgG1 titers. As reported previously using polystyrene nanoparticles (NPs), p.o.-primed mice developed high titers of IgG2c and intestinal IgA following s.c. boosting immunization with LP-Ova. Mice that received a single s.c. immunization with LP-Ova did not develop serum IgG2c or intestinal IgA antibodies. Additionally, in s.c.-immunized mice serum IgG1 titers decreased significantly by 3 months after immunization. In contrast, in mice primed p.o. and boosted s.c. with LP-Ova, serum IgG1/IgG2c, and intestinal IgA antibody titers remained stable. Administration of LPs exerted no adverse effects on immunized mice as no morbidity or signs of toxicity were observed for the duration of the studies. These results indicate that antigen-conjugated liposomes are immunogenic and confirm a previous report that mucosal priming followed by a s.c. boosting immunization is the most effective strategy for inducing long-lasting mucosal IgA, as well as a polarized Th1/Th2 systemic response. In addition to being biodegradable and easily functionalized by conjugation, liposomes have a hollow core which can also be loaded with cargo, allowing for a targeted delivery of multiple antigens (or drugs) simultaneously. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 557-565, 2017.


Assuntos
Antígenos , Imunidade nas Mucosas/efeitos dos fármacos , Imunogenicidade da Vacina/imunologia , Nanopartículas/química , Polímeros , Poliestirenos , Poli-Inos , Animais , Antígenos/química , Antígenos/imunologia , Antígenos/farmacologia , Imunização Secundária , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Lipossomos , Camundongos , Polímero Poliacetilênico , Polímeros/química , Polímeros/farmacologia , Poliestirenos/química , Poliestirenos/imunologia , Poliestirenos/farmacologia , Poli-Inos/química , Poli-Inos/imunologia , Poli-Inos/farmacologia
6.
Curr Med Chem ; 23(6): 578-88, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26639094

RESUMO

Janus kinase-2 (JAK2) is a non-receptor tyrosine kinase signaling molecule that mediates the effects of various hormones and cytokines, including interferon, erythropoietin, leptin, and growth hormone. It also fosters tumor growth and modifies the activity of several nutrient transporters. JAK2 contributes to the regulation of the cell volume, protectS cells during energy depletion, proliferation, and aids the survival of tumor cells. Recently, JAK2 was identified as a powerful regulator of transport processes across the plasma membrane. Either directly or indirectly JAK2 may stimulate or inhibit transporter proteins, including ion channels, carriers and Na(+)/K(+) pumps. As a powerful regulator of transport mechanisms across the cell membrane, JAK2 regulates a wide variety of potassium, calcium, sodium and chloride ion channels, multiple Na+-coupled cellular carriers including EAAT1-4, NaPi-IIa, SGLT1, BoaT1, PepT1-2, CreaT1, SMIT1, and BGT1 as well as Na(+)/K(+)-ATPase. These cellular transport regulations contribute to various physiological and pathophysiological processes and thus exerting JAK2-sensitive effects. Future investigations will be important to determine whether JAK2 regulates cell-surface expression of other transporters and further elucidate underlying mechanisms governing JAK2 actions.


Assuntos
Proteínas de Transporte/metabolismo , Janus Quinase 2/metabolismo , Animais , Transporte Biológico , Humanos , Canais Iônicos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
7.
Antibodies (Basel) ; 5(4)2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31558001

RESUMO

Nanoparticles (NPs) are increasingly being used for drug delivery, as well as antigen carriers and immunostimulants for the purpose of developing vaccines. In this work, we examined how intranasal (i.n.) priming followed by i.n. or subcutaneous (s.c.) boosting immunization affects the humoral immune response to chicken ovalbumin (Ova) and Ova conjugated to 20 nm NPs (NP-Ova). We show that i.n. priming with 20 mg of soluble Ova, a dose known to trigger oral tolerance when administered via gastric gavage, induced substantial systemic IgG1 and IgG2c, as well as mucosal antibodies. These responses were further boosted following a s.c. immunization with Ova and complete Freund's adjuvant (Ova+CFA). In contrast, 100 µg of Ova delivered via NPs induced an IgG1-dominated systemic response, and primed the intestinal mucosa for secretion of IgA. Following a secondary s.c. or i.n. immunization with Ova+CFA or NP-Ova, systemic IgG1 titers significantly increased, and serum IgG2c and intestinal antibodies were induced in mice primed nasally with NP-Ova. Only Ova- and NP-Ova-primed mice that were s.c.-boosted exhibited substantial systemic and mucosal titers for up to 6 months after priming, whereas the antibodies of i.n.-boosted mice declined over time. Our results indicate that although the amount of Ova delivered by NPs was 1000-fold less than Ova delivered in soluble form, the antigen-specific antibody responses, both systemic and mucosal, are essentially identical by 6 months following the initial priming immunization. Additionally, both i.n.- and s.c.-boosting strategies for NP-Ova-primed mice were capable of inducing a polarized Th1/Th2 immune response, as well as intestinal antibodies; however, it is only by using a heterogeneous prime-boost strategy that long-lasting antibody responses were initiated. These results provide valuable insight for future mucosal vaccine development, as well as furthering our understanding of mucosal antibody responses.

8.
PLoS One ; 10(6): e0130092, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26068006

RESUMO

Infection of mice with Salmonella enterica serovar Typhimurium (Salmonella) causes systemic inflammatory disease and enlargement of the spleen (splenomegaly). Splenomegaly has been attributed to a general increase in the numbers of phagocytes, lymphocytes, as well as to the expansion of immature CD71+Ter119+ reticulocytes. The spleen is important for recycling senescent red blood cells (RBCs) and for the capture and eradication of blood-borne pathogens. Conservation of splenic tissue architecture, comprised of the white pulp (WP), marginal zone (MZ), and red pulp (RP) is essential for initiation of adaptive immune responses to captured pathogens. Using flow cytometry and four color immunofluorescence microscopy (IFM), we show that Salmonella-induced splenomegaly is characterized by drastic alterations of the splenic tissue architecture and cell population proportions, as well as in situ cell distributions. A major cause of splenomegaly appears to be the significant increase in immature RBC precursors and F4/80+ macrophages that are important for recycling of heme-associated iron. In contrast, the proportions of B220+, CD4+ and CD8+ lymphocytes, as well as MZ MOMA+ macrophages decrease significantly as infection progresses. Spleen tissue sections show visible tears and significantly altered tissue architecture with F4/80+ macrophages and RBCs expanding beyond the RP and taking over most of the spleen tissue. Additionally, F4/80+ macrophages actively phagocytose not only RBCs, but also lymphocytes, indicating that they may contribute to declining lymphocyte proportions during Salmonella infection. Understanding how these alterations of spleen microarchitecture impact the generation of adaptive immune responses to Salmonella has implications for understanding Salmonella pathogenesis and for the design of more effective Salmonella-based vaccines.


Assuntos
Subpopulações de Linfócitos B/patologia , Macrófagos/patologia , Infecções por Salmonella/patologia , Baço/patologia , Subpopulações de Linfócitos T/patologia , Animais , Subpopulações de Linfócitos B/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Infecções por Salmonella/sangue , Infecções por Salmonella/imunologia , Salmonella typhimurium , Baço/imunologia , Esplenomegalia , Subpopulações de Linfócitos T/imunologia
9.
PLoS One ; 10(2): e0118067, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25710518

RESUMO

Food or water-borne enteric pathogens invade their hosts via intestinal mucosal surfaces, thus developing effective oral vaccines would greatly reduce the burden of infectious diseases. The nature of the antigen, as well as the mode of its internalization in the intestinal mucosa affects the ensuing immune response. We show that model protein antigen ovalbumin (Ova) given per-orally (p.o.) induces oral tolerance (OT), characterized by systemic IgG1-dominated antibody response, which cannot be boosted by sub-cutaneous (s.c.) immunization with Ova in complete Freund's adjuvant (CFA). Intestinal IgA generated in response to Ova feeding diminished over time and was abrogated by s.c. immunization with Ova+CFA. Humoral response to Ova was altered by administering Ova conjugated to 20 nm nanoparticles (NP-Ova). P.o. administration of NP-Ova induced systemic IgG1/IgG2c, and primed the intestinal mucosa for secretion of IgA. These responses were boosted by secondary s.c. immunization with Ova+CFA or p.o. immunization with NP-Ova. However, only in s.c.-boosted mice serum and mucosal antibody titers remained elevated for 6 months after priming. In contrast, s.c. priming with NP-Ova induced IgG1-dominated serum antibodies, but did not prime the intestinal mucosa for secretion of IgA, even after secondary p.o. immunization with NP-Ova. These results indicate that Ova conjugated to NPs reaches the internal milieu in an immunogenic form and that mucosal immunization with NP-Ova is necessary for induction of a polarized Th1/Th2 immune response, as well as intestinal IgA response. In addition, mucosal priming with NP-Ova, followed by s.c. boosting induces superior systemic and mucosal memory responses. These findings are important for the development of efficacious mucosal vaccines.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Antígenos/farmacologia , Imunidade nas Mucosas/efeitos dos fármacos , Nanopartículas/química , Ovalbumina/farmacologia , Administração Oral , Animais , Antígenos/administração & dosagem , Antígenos/química , Ensaio de Imunoadsorção Enzimática , Feminino , Adjuvante de Freund/imunologia , Imunização Secundária , Imunoglobulina A/metabolismo , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Injeções Subcutâneas , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Ovalbumina/química , Tamanho da Partícula , Células Th1/imunologia , Células Th2/imunologia
10.
PLoS One ; 9(12): e114601, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25490456

RESUMO

The female reproductive tract (FRT) includes the oviducts (fallopian tubes), uterus, cervix and vagina. A layer of columnar epithelium separates the endocervix and uterus from the outside environment, while the vagina is lined with stratified squamous epithelium. The mucosa of the FRT is exposed to antigens originating from microflora, and occasionally from infectious microorganisms. Whether epithelial cells (ECs) of the FRT take up (sample) the lumen antigens is not known. To address this question, we examined the uptake of 20-40 nm nanoparticles (NPs) applied vaginally to mice which were not treated with hormones, epithelial disruptors, or adjuvants. We found that 20 and 40 nm NPs are quickly internalized by ECs of the upper FRT and within one hour could be observed in the lymphatic ducts that drain the FRT, as well as in the ileac lymph nodes (ILNs) and the mesenteric lymph nodes (MLNs). Chicken ovalbumin (Ova) conjugated to 20 nm NPs (NP-Ova) when administered vaginally reaches the internal milieu in an immunologically relevant form; thus vaginal immunization of mice with NP-Ova induces systemic IgG to Ova antigen. Most importantly, vaginal immunization primes the intestinal mucosa for secretion of sIgA. Sub-cutaneous (s.c) boosting immunization with Ova in complete Freund's adjuvant (CFA) further elevates the systemic (IgG1 and IgG2c) as well as mucosal (IgG1 and sIgA) antibody titers. These findings suggest that the modes of antigen uptake at mucosal surfaces and pathways of antigen transport are more complex than previously appreciated.


Assuntos
Antígenos/metabolismo , Células Epiteliais/metabolismo , Nanopartículas/análise , Absorção Vaginal , Animais , Antígenos/imunologia , Transporte Biológico , Células Epiteliais/imunologia , Feminino , Imunidade nas Mucosas , Imunização/métodos , Imunoglobulina G/sangue , Linfonodos/imunologia , Linfonodos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia
11.
PLoS One ; 9(1): e86656, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475164

RESUMO

Intestinal epithelial cells (IECs) overlying the villi play a prominent role in absorption of digested nutrients and establish a barrier that separates the internal milieu from potentially harmful microbial antigens. Several mechanisms by which antigens of dietary and microbial origin enter the body have been identified; however whether IECs play a role in antigen uptake is not known. Using in vivo imaging of the mouse small intestine, we investigated whether epithelial cells (enterocytes) play an active role in the uptake (sampling) of lumen antigens. We found that small molecular weight antigens such as chicken ovalbumin, dextran, and bacterial LPS enter the lamina propria, the loose connective tissue which lies beneath the epithelium via goblet cell associated passageways. However, epithelial cells overlying the villi can internalize particulate antigens such as bacterial cell debris and inert nanoparticles (NPs), which are then found co-localizing with the CD11c+ dendritic cells in the lamina propria. The extent of NP uptake by IECs depends on their size: 20-40 nm NPs are taken up readily, while NPs larger than 100 nm are taken up mainly by the epithelial cells overlying Peyer's patches. Blocking NPs with small proteins or conjugating them with ovalbumin does not inhibit their uptake. However, the uptake of 40 nm NPs can be inhibited when they are administered with an endocytosis inhibitor (chlorpromazine). Delineating the mechanisms of antigen uptake in the gut is essential for understanding how tolerance and immunity to lumen antigens are generated, and for the development of mucosal vaccines and therapies.


Assuntos
Antígenos/metabolismo , Enterócitos/metabolismo , Intestino Delgado/citologia , Absorção , Animais , Western Blotting , Dextranos , Intestino Delgado/metabolismo , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Microscopia de Fluorescência , Ovalbumina , Tamanho da Partícula
12.
Nature ; 483(7389): 345-9, 2012 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-22422267

RESUMO

The intestinal immune system is exposed to a mixture of foreign antigens from diet, commensal flora and potential pathogens. Understanding how pathogen-specific immunity is elicited while avoiding inappropriate responses to the background of innocuous antigens is essential for understanding and treating intestinal infections and inflammatory diseases. The ingestion of protein antigen can induce oral tolerance, which is mediated in part by a subset of intestinal dendritic cells (DCs) that promote the development of regulatory T cells. The lamina propria (LP) underlies the expansive single-cell absorptive villous epithelium and contains a large population of DCs (CD11c(+) CD11b(+) MHCII(+) cells) comprised of two predominant subsets: CD103(+) CX(3)CR1(-) DCs, which promote IgA production, imprint gut homing on lymphocytes and induce the development of regulatory T cells, and CD103(-) CX(3)CR1(+) DCs (with features of macrophages), which promote tumour necrosis factor-α (TNF-α) production, colitis, and the development of T(H)17 T cells. However, the mechanisms by which different intestinal LP-DC subsets capture luminal antigens in vivo remains largely unexplored. Using a minimally disruptive in vivo imaging approach we show that in the steady state, small intestine goblet cells (GCs) function as passages delivering low molecular weight soluble antigens from the intestinal lumen to underlying CD103(+) LP-DCs. The preferential delivery of antigens to DCs with tolerogenic properties implies a key role for this GC function in intestinal immune homeostasis.


Assuntos
Antígenos CD/metabolismo , Antígenos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Caliciformes/imunologia , Tolerância Imunológica/imunologia , Cadeias alfa de Integrinas/metabolismo , Intestino Delgado/imunologia , Animais , Antígenos/metabolismo , Células Dendríticas/citologia , Dieta , Células Caliciformes/metabolismo , Homeostase , Humanos , Imunoglobulina A/imunologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Solubilidade , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/imunologia
13.
Immunity ; 33(4): 597-606, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-20933441

RESUMO

Dendritic cells (DCs) comprise distinct functional subsets including CD8⁻ and CD8(+) classical DCs (cDCs) and interferon-secreting plasmacytoid DCs (pDCs). The cytokine Flt3 ligand (Flt3L) controls the development of DCs and is particularly important for the pDC and CD8(+) cDC and their CD103(+) tissue counterparts. We report that mammalian target of rapamycin (mTOR) inhibitor rapamycin impaired Flt3L-driven DC development in vitro, with the pDCs and CD8(+)-like cDCs most profoundly affected. Conversely, deletion of the phosphoinositide 3-kinase (PI3K)-mTOR negative regulator Pten facilitated Flt3L-driven DC development in culture. DC-specific Pten targeting in vivo caused the expansion of CD8(+) and CD103(+) cDC numbers, which was reversible by rapamycin. The increased CD8(+) cDC numbers caused by Pten deletion correlated with increased susceptibility to the intracellular pathogen Listeria. Thus, PI3K-mTOR signaling downstream of Flt3L controls DC development, and its restriction by Pten ensures optimal DC pool size and subset composition.


Assuntos
Células Dendríticas/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/fisiologia , Animais , Antígenos CD/análise , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Cadeias alfa de Integrinas/análise , Listeriose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , PTEN Fosfo-Hidrolase/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
14.
Avian Dis ; 53(2): 149-55, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19630217

RESUMO

Marek's disease virus (MDV) is an acute transforming alphaherpesvirus of chickens that causes Marek's disease. During the infection of chickens, MDV establishes latency in CD4+ (T-helper) cells, which are also the target of transformation. The study of MDV latency has been limited to the use of MDV tumor-derived cell lines or blood cells isolated from chickens during presumed periods of latent infection. In 1992 Pratt et al. described the uptake of the MDV genome by a reticuloendotheliosis-transformed T-cell line (RECC-CU91). They reported that MDV established latency in CU91 cells, but that MDV genome expression was very limited. In this report we have examined the uptake of oncogenic, recombinant, and vaccine strain MDVs. We report that the entire MDV genome is taken up by CU91 cells, is hypomethylated, and readily reactivates from this latent state in a manner similar to MDV-transformed cell lines. Notably, virus could not be recovered from cell lines harboring vaccine virus CVI988 or the JM102 strain of MDV. Overall these cell lines present a useful model for the further study of MDV latency, particularly for those viruses having mutations that may affect replication or fitness of the virus in vivo. In addition, these cell lines offer an attractive means to study the latency of vaccine viruses, which establish relatively low levels of latent infection in vivo.


Assuntos
Mardivirus/fisiologia , Linfócitos T/virologia , Latência Viral/fisiologia , Animais , Linhagem Celular , Transformação Celular Viral , Galinhas , Genoma Viral , Vírus da Reticuloendoteliose Aviária
15.
Avian Dis ; 53(2): 156-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19630218

RESUMO

Marek's disease virus (MDV) is an alphaherpesvirus of chickens that causes the paralysis and rapid lymphoma formation known as Marek's disease. MDV establishes latent infection in activated CD4+ T-cells, and these cells are also the target for transformation. MDV latency has been studied using MDV lymphoma-derived cell lines and T-cells isolated from infected chickens. Each of these models has limitations because MDV-transformed cell lines require the use of oncogenic viruses; conversely, pools of latently infected cells are in relatively low abundance and invariably contain cells undergoing reactivation to lytic infection. In this study we have examined the spontaneous and induced expression of the MDV genome, the effect of genome uptake on cellular proliferation and apoptosis resistance, and differences in cellular surface antigen expression associated with MDV genome uptake in a reticuloendotheliosis virus (REV)-transformed T-cell model. We report that the MDV genome is highly transcribed during this latent infection, and that the expression of Marek's EcoRI-Q-encoded protein (Meq) transcripts is similar to that of MDV-transformed cells, but is somewhat lower than MDV-transformed cells at the protein level. Uptake of the MDV genome was associated with an increased growth rate and resistance to serum starvation-induced apoptosis. Treatment of cells with bromodeoxyuridine induced the expression of MDV lyric antigens in a manner similar to MDV-transformed cells. Uptake of the MDV genome, however, was not consistently associated with alteration ofT-cell surface antigen expression. Overall, our data show that the REV-transformed cell line model for MDV latency mimics many important aspects of latency also observed in MDV-transformed cells and provides an additional tool for examining MDV latent infection.


Assuntos
Regulação Viral da Expressão Gênica/fisiologia , Mardivirus/fisiologia , Linfócitos T/virologia , Latência Viral/fisiologia , Animais , Linhagem Celular , Transformação Celular Viral , Galinhas , Genoma Viral , Vírus da Reticuloendoteliose Aviária
16.
Curr Top Microbiol Immunol ; 334: 199-226, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19521687

RESUMO

Listeria monocytogenes infection in mice is a highly prolific model of bacterial infection. Several in vivo imaging approaches have been used to study host cell dynamics in response to infection, including bioluminescence imaging, confocal microscopy and two-photon microscopy, The application of in vivo imaging to study transgenic mouse models is providing unprecedented opportunities to test specific molecular mechanistic theories about how the host immune response unfolds. In complementary studies, in vivo imaging can be performed using genetically engineered bacterial mutants to assess the impact of specific virulence factors in host cell invasion and pathogenesis. The purpose of this chapter is to provide a general rationale for why in vivo imaging is important, provide an overview of various techniques highlighting the strengths and weaknesses of each, and provide examples of how various imaging techniques have been used to study Listeria infection. Lastly, our goal is to make the reader aware of the tremendous potential these approaches hold for studying host-pathogen interactions.


Assuntos
Interações Hospedeiro-Patógeno , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Microscopia Confocal , Animais , Modelos Animais de Doenças , Corantes Fluorescentes/metabolismo , Humanos , Listeria monocytogenes/imunologia , Listeriose/microbiologia , Listeriose/patologia , Camundongos , Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Virulência
17.
Eur J Immunol ; 39(2): 417-25, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19130474

RESUMO

The spleen is an important organ for the host response to systemic bacterial infections. Many cell types and cell surface receptors have been shown to play role in the capture and control of bacteria, yet these are often studied individually and a coherent picture has yet to emerge of how various phagocytes collaborate to control bacterial infection. We analyzed the cellular distribution of Listeria monocytogenes (LM) in situ during the early phase of infection. Using an immunohistochemistry approach, five distinct phagocyte populations contained LM after i.v. challenge and accounted for roughly all bacterial signal in tissue sections. Our analysis showed that LM was initially captured by a wide range of phagocytes in the marginal zone, where the growth of LM appeared to be controlled. The cellular distribution of LM within phagocyte populations changed rapidly during the first few hours, decreasing in marginal zone macrophages and transiently increasing in CD11c(+) DC. After 4-6 h LM was transported to the periarteriolar lymphoid sheath where the infective foci developed and LM grew exponentially.


Assuntos
Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Fagócitos/microbiologia , Baço/microbiologia , Animais , Listeria monocytogenes/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Fagócitos/imunologia , Baço/imunologia
18.
Cell Microbiol ; 11(4): 551-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19170686

RESUMO

Two-photon (2P) microscopy has become increasingly popular among immunologists for analysing single-cell dynamics in tissues. Researchers are now taking 2P microscopy beyond the study of model antigen systems (e.g. ovalbumin immunization) and are applying the technique to examine infection in vivo. With the appropriate fluorescent probes, 2P imaging can provide high-resolution spatio-temporal information regarding cell behaviour, monitor cell functions and assess various outcomes of infection, such as host cell apoptosis or pathogen proliferation. Imaging of transgenic and knockout mice can be used to probe molecular mechanisms governing the host response to infection. From the microbe side, imaging genetically engineered mutant strains of a pathogen can test the roles of specific virulence factors in pathogenesis. Here, we discuss recent work that has applied 2P microscopy to study models of infection and highlight the tremendous potential that this approach has for investigating host-pathogen interactions.


Assuntos
Infecções Bacterianas/patologia , Interações Hospedeiro-Patógeno , Microscopia/métodos , Fótons , Infecções por Protozoários/patologia , Viroses/patologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Modelos Animais de Doenças , Eucariotos/fisiologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Infecções por Protozoários/imunologia , Infecções por Protozoários/parasitologia , Viroses/imunologia , Viroses/virologia , Vírus/patogenicidade
19.
Immunity ; 29(3): 476-86, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18760639

RESUMO

The spleen plays an important role in host-protective responses to bacteria. However, the cellular dynamics that lead to pathogen-specific immunity remain poorly understood. Here we examined Listeria monocytogenes (Lm) infection in the mouse spleen via in situ fluorescence microscopy. We found that the redistribution of Lm from the marginal zone (MZ) to the periarteriolar lymphoid sheath (PALS) was inhibited by pertussis toxin and required the presence of CD11c(+) cells. As early as 9 hr after infection, we detected infected dendritic cells in the peripheral regions of the PALS and clustering of Lm-specific T cells by two-photon microscopy. Pertussis toxin inhibited both Lm entry into the PALS and antigen presentation to CD8(+) T cells. Our study suggests that splenic dendritic cells rapidly deliver intracellular bacteria to the T cell areas of the white pulp to initiate CD8(+) T cell responses.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Listeriose/imunologia , Baço/imunologia , Baço/microbiologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Ácido Clodrônico/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Lipossomos , Listeria monocytogenes/imunologia , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Toxina Pertussis/farmacologia , Baço/citologia , Baço/efeitos dos fármacos
20.
Infect Immun ; 76(12): 5745-53, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18809658

RESUMO

Recombinant attenuated Salmonella vaccines against avian coccidiosis were developed to deliver Eimeria species antigens to the lymphoid tissues of chickens via the type 3 secretion system (T3SS) and the type 2 secretion system (T2SS) of Salmonella. For antigen delivery via the T3SS, the Eimeria tenella gene encoding sporozoite antigen SO7 was cloned downstream of the translocation domain of the Salmonella enterica serovar Typhimurium sopE gene in the parental pYA3868 and pYA3870 vectors to generate pYA4156 and pYA4157. Newly constructed T3SS vectors were introduced into host strain chi8879 (Delta phoP233 Delta sptP1033::xylE Delta asdA16), an attenuated derivative of the highly virulent UK-1 strain. The SopE-SO7 fusion protein was secreted by the T3SS of Salmonella. The vector pYA4184 was constructed for delivery of the SO7 antigen via the T2SS. The SO7 protein was toxic to Salmonella when larger amounts were synthesized; thus, the synthesis of this protein was placed under the control of the lacI repressor gene, whose expression in turn was dependent on the amount of available arabinose in the medium. The pYA4184 vector was introduced into host strain chi9242 (Delta phoP233 Delta asdA16 Delta araBAD23 Delta relA198::araC P(BAD) lacI TT [TT is the T4ipIII transcription terminator]). In addition to SO7, for immunization and challenge studies we used the EAMZ250 antigen of Eimeria acervulina, which was previously shown to confer partial protection against E. acervulina challenge when it was delivered via the T3SS. Immunization of chickens with a combination of the SO7 and EAMZ250 antigens delivered via the T3SS induced superior protection against challenge by E. acervulina. In contrast, chickens immunized with SO7 that was delivered via the T2SS of Salmonella were better protected from challenge by E. tenella.


Assuntos
Antígenos de Protozoários/imunologia , Coccidiose/prevenção & controle , Vacinas Protozoárias/imunologia , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia , Animais , Antígenos de Protozoários/biossíntese , Antígenos de Protozoários/genética , Western Blotting , Galinhas , Coccidiose/veterinária , Eimeria tenella/genética , Vetores Genéticos , Masculino , Plasmídeos/genética , Salmonella typhimurium/genética , Vacinas Atenuadas/genética , Vacinas Sintéticas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...