Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Intervalo de año de publicación
1.
Fish Shellfish Immunol ; 121: 62-73, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34998096

RESUMEN

The X-organ-sinus gland complex (XO-SG) in the eyestalk is an important neuroendocrine regulatory organ of crustaceans such as Litopenaeus vannamei, a prominent aquaculture species. The current study found significant changes in the enzyme activities of ALP, ACP, and T-SOD of hepatopancreatic in response to Vibrio parahaemolyticus exposure following eyestalk ablation, indicating that they were all involved in the immunological regulation of shrimps against V. parahaemolyticus infection. A total of 52,656 unigenes were obtained after RNA-Seq, with an average length of 1036 bp and an N50 of 1847 bp. Subsequently, 1899 eyestalk positive regulation genes (EPRGs), 745 eyestalk negative regulation genes (ENRGs), and 2077 non-eyestalk regulatory genes (NEGs) were identified. KEGG analysis of EPRGs revealed that eyestalk ablation might activate the neuroendocrine-immune (NEI) system. The RNA-Seq data were validated using quantitative real-time PCR (qRT-PCR). The findings suggested that eyestalk ablation might affect the expression of genes involved in the prophenoloxidase-activating system, the TLR signaling pathway, and numerous other immune-related genes in L. vannamei. All of these findings revealed that the eyestalk might have a role in the immune response of L. vannamei. The genes and pathways discovered in this study will help to elucidate the molecular mechanisms of hemocytes' immune response to V. parahaemolyticus following eyestalk ablation in shrimp, as well as provide the framework for building crustacean immunity theory.


Asunto(s)
Estructuras Animales/inmunología , Inmunidad Innata , Penaeidae , Vibriosis , Animales , Acuicultura , Hemocitos , Penaeidae/genética , Penaeidae/inmunología , Vibrio parahaemolyticus
2.
Front Immunol ; 12: 769775, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804060

RESUMEN

The crosstalk between the immune system and microbiota drives an amazingly complex mutualistic symbiosis. In mammals, the upper respiratory tract acts as a gateway for pathogen invasion, and the dynamic interaction between microbiota and mucosal immunity on its surface can effectively prevent disease development. However, the relationship between virus-mediated mucosal immune responses and microbes in lower vertebrates remains uncharacterized. In this study, we successfully constructed an infection model by intraperitoneally injecting common carp (Cyprinus carpio) with spring viremia of carp virus (SVCV). In addition to the detection of the SVCV in the nose and pharynx of common carp, we also identified obvious histopathological changes following viral infection. Moreover, numerous immune-related genes were significantly upregulated in the nose and pharynx at the peak of SVCV infection, after which the expression levels decreased to levels similar to those of the control group. Transcriptome sequencing results revealed that pathways associated with bacterial infection in the Toll-like receptor pathway and the Nod-like receptor pathway were activated in addition to the virus-related Rig-I-like receptor pathway after SVCV infection, suggesting that viral infection may be followed by opportunistic bacterial infection in these mucosal tissues. Using 16S rRNA gene sequencing, we further identified an upward trend in pathogenic bacteria on the mucosal surface of the nose and pharynx 4 days after SVCV infection, after which these tissues eventually reached new homeostasis. Taken together, our results suggest that the dynamic interaction between mucosal immunity and microbiota promotes the host to a new ecological state.


Asunto(s)
Bacterias/inmunología , Carpas/inmunología , Enfermedades de los Peces/inmunología , Inmunidad Mucosa/inmunología , Faringe/inmunología , Rhabdoviridae/inmunología , Estructuras Animales/inmunología , Estructuras Animales/microbiología , Estructuras Animales/virología , Animales , Bacterias/clasificación , Bacterias/genética , Carpas/microbiología , Carpas/virología , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/métodos , Homeostasis/genética , Homeostasis/inmunología , Inmunidad Mucosa/genética , Faringe/microbiología , Faringe/virología , Filogenia , ARN Ribosómico 16S/genética , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/inmunología , Rhabdoviridae/genética , Rhabdoviridae/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología
3.
Vet Immunol Immunopathol ; 230: 110129, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33059181

RESUMEN

BACKGROUND: In order to adequately monitor cytokines in experimental models, currently available methods and commercially available kits should be compared. AIM: To compare the plasma and tissue concentrations of IL-1ß, IL-6, IL-8, IL-10, and TNF as a measure of systemic inflammation in septic pigs. METHODS: Cytokines were quantified from blood and tissue samples obtained at 0, 60, 120, 180, and 240 min, and in postmortem biopsies of the liver, kidney, lung, heart, and spleen from 26 anesthetized landrace pigs. (24 with experimental sepsis, two sham controls). Porcine-specific ELISAs (R&D) and multiplex (9-plex from Thermo Fischer, 13-plex from Millipore) immunoassays were compared. RESULTS: The assays differed for the different cytokines and between blood and tissue. In blood, the highest concentration of TNF and IL-6 was in ELISA, IL-1ß equal in ELISA and 13-plex, IL-8 in 13-plex and IL-10 in 9-plex. In tissue, the highest concentration of TNF and IL-1ß was in ELISA, IL-6 and IL-8 in 13-plex and IL-10 in 9-plex. CONCLUSION: The choice of analysis impacts the quantified cytokine responses in porcine models. ELISA and multiplex techniques supplement each other and our data suggest which assays to use for the quantification of the different cytokines.


Asunto(s)
Estructuras Animales/inmunología , Citocinas/sangre , Inmunoensayo/normas , Inmunoensayo/veterinaria , Inflamación/veterinaria , Sepsis/veterinaria , Enfermedades de los Porcinos/inmunología , Animales , Citocinas/clasificación , Ensayo de Inmunoadsorción Enzimática/normas , Inmunoensayo/métodos , Juego de Reactivos para Diagnóstico/normas , Porcinos
4.
Theranostics ; 10(14): 6430-6447, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32483462

RESUMEN

Rationale: Zika virus (ZIKV) is a pathogenic virus known to cause a wide range of congenital abnormalities, including microcephaly, Guillain-Barre syndrome, meningoencephalitis, and other neurological complications, in humans. This study investigated the noninvasive detection of ZIKV infection in vivo, which is necessary for elucidating the virus's mechanisms of viral replication and pathogenesis, as well as to accelerate the development of anti-ZIKV therapeutic strategies. Methods: In this study, a recombinant ZIKV harbouring Nluc gene (ZIKV-Nluc) was designed, recovered, and purified. The levels of bioluminescence were directly correlated with viral loads in vitro and in vivo. The dynamics of ZIKV infection in A129 (interferon (IFN)-α/ß receptor deficient), AG6 (IFN-α/ß and IFN-γ receptor deficient), and C57BL/6 mice were characterized. Pregnant dams were infected with ZIKV-Nluc at E10 via intra footpad injection. Then, the pooled immune sera (anti-ZIKV neutralizing antibodies) #22-1 in ZIKV-Nluc virus-infected mice were visualized. Results: ZIKV-Nluc showed a high genetic stability and replicated well in cells with similar properties to the wild-type ZIKV (ZIKVwt). Striking bioluminescence signals were consistently observed in animal organs, including spleen, intestine, testis, uterus/ovary, and kidney. The ileocecal junction was found to be the crucial visceral target. Infection of pregnant dams with ZIKV-Nluc showed that ZIKV was capable of crossing the maternal-fetal barrier to infect the fetuses via vertical transmission. Furthermore, it was visualized that treatment with the pooled immune sera was found to greatly restrict the spread of the ZIKV-Nluc virus in mice. Conclusions: This study is the first to report the real-time noninvasive tracking of the progression of ZIKV invading immune-sheltered tissues and propagating vertically during pregnancy. The results demonstrate that ZIKV-Nluc represents a powerful tool for the study of the replication, dissemination, pathogenesis, and treatment of ZIKV in vitro and in vivo.


Asunto(s)
Mediciones Luminiscentes/métodos , Infección por el Virus Zika/diagnóstico por imagen , Estructuras Animales/diagnóstico por imagen , Estructuras Animales/inmunología , Estructuras Animales/virología , Animales , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , Modelos Animales de Enfermedad , Femenino , Feto/diagnóstico por imagen , Feto/virología , Transmisión Vertical de Enfermedad Infecciosa , Ratones , Ratones Endogámicos C57BL/virología , Embarazo , Carga Viral/efectos de los fármacos , Carga Viral/inmunología , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología , Virus Zika/efectos de los fármacos , Infección por el Virus Zika/inmunología
5.
Vaccine ; 35(33): 4184-4196, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28662952

RESUMEN

The protozoan parasite Histomonas meleagridis is the causative agent of histomonosis in gallinaceous birds. In turkeys, the disease can result in high mortality due to severe inflammation and necrosis in caecum and liver, whereas in chickens the disease is less severe. Recently, experimental vaccination was shown to protect chickens and turkeys against histomonosis but dynamics in the cellular immune response are not yet demonstrated. In the present work, different groups of birds of both species were vaccinated with attenuated, and/or infected with virulent histomonads. Flow cytometry was applied at different days post inoculation to analyse the absolute number of T-cell subsets and B cells in caecum, liver, spleen and blood, in order to monitor changes in these major lymphocyte subsets. In addition, in chicken samples total white blood cells were investigated. Infected turkeys showed a significant decrease of T cells in the caecum within one week post infection compared to control birds, whereas vaccination showed delayed changes. The challenge of vaccinated turkeys led to a significant increase of all investigated lymphocytes in the blood already at 4 DPI, indicating an effective and fast recall response of the primed immune system. In the caecum of chickens, changes of B cells, CD4+ and CD8α+ T cells were much less pronounced than in turkeys, however, mostly caused by virulent histomonads. Analyses of whole blood in non-vaccinated but infected chickens revealed increasing numbers of monocytes/macrophages on all sampling days, whereas a decrease of heterophils was observed directly after challenge, suggesting recruitment of this cell population to the local site of infection. Our results showed that virulent histomonads caused more severe changes in the distribution of lymphocyte subsets in turkeys compared to chickens. Moreover, vaccination with attenuated histomonads resulted in less pronounced alterations in both species, even after challenge.


Asunto(s)
Linfocitos B/inmunología , Enfermedades de las Aves de Corral/prevención & control , Infecciones por Protozoos/prevención & control , Vacunas Antiprotozoos/inmunología , Subgrupos de Linfocitos T/inmunología , Estructuras Animales/inmunología , Animales , Sangre/inmunología , Pollos , Citometría de Flujo , Infecciones por Protozoos/inmunología , Infecciones por Protozoos/patología , Vacunas Antiprotozoos/administración & dosificación , Pavos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
6.
Viruses ; 8(10)2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27763552

RESUMEN

Rodent-borne hantaviruses can cause two human diseases with many pathological similarities: hantavirus cardiopulmonary syndrome (HCPS) in the western hemisphere and hemorrhagic fever with renal syndrome in the eastern hemisphere. Each virus is hosted by specific reservoir species without conspicuous disease. HCPS-causing hantaviruses require animal biosafety level-4 (ABSL-4) containment, which substantially limits experimental research of interactions between the viruses and their reservoir hosts. Maporal virus (MAPV) is a South American hantavirus not known to cause disease in humans, thus it can be manipulated under ABSL-3 conditions. The aim of this study was to develop an ABSL-3 hantavirus infection model using the deer mouse (Peromyscus maniculatus), the natural reservoir host of Sin Nombre virus (SNV), and a virus that is pathogenic in another animal model to examine immune response of a reservoir host species. Deer mice were inoculated with MAPV, and viral RNA was detected in several organs of all deer mice during the 56 day experiment. Infected animals generated both nucleocapsid-specific and neutralizing antibodies. Histopathological lesions were minimal to mild with the peak of the lesions detected at 7-14 days postinfection, mainly in the lungs, heart, and liver. Low to modest levels of cytokine gene expression were detected in spleens and lungs of infected deer mice, and deer mouse primary pulmonary cells generated with endothelial cell growth factors were susceptible to MAPV with viral RNA accumulating in the cellular fraction compared to infected Vero cells. Most features resembled that of SNV infection of deer mice, suggesting this model may be an ABSL-3 surrogate for studying the host response of a New World hantavirus reservoir.


Asunto(s)
Inmunidad Adaptativa , Modelos Animales de Enfermedad , Infecciones por Hantavirus/patología , Infecciones por Hantavirus/virología , Orthohantavirus/inmunología , Orthohantavirus/patogenicidad , Peromyscus/virología , Estructuras Animales/inmunología , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Citocinas/biosíntesis , Histocitoquímica , ARN Viral/análisis , Factores de Tiempo
7.
Sci Rep ; 5: 16597, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26593439

RESUMEN

Bathymodiolid mussels dominate hydrothermal vents, cold methane/sulfide-hydrocarbon seeps, and other sites of organic enrichment. Here, we aimed to explore the innate immune system and detoxification mechanism of the deep sea mussel Bathymodiolus platifrons collected from a methane seep in the South China Sea. We sequenced the transcriptome of the mussels' gill, foot and mantle tissues and generated a transcriptomic database containing 96,683 transcript sequences. Based on GO and KEGG annotations, we reported transcripts that were related to the innate immune system, heavy metal detoxification and sulfide metabolic genes. Our in-depth analysis on the isoforms of peptidoglycan recognition protein (PGRP) that have different cellular location and potentially differential selectivity towards peptidoglycan (PGN) from gram-positive and gram-negative bacteria were differentially expressed in different tissues. We also reported a potentially novel form of metallothionein and the production of phytochelatin in B. platifrons, which has not been reported in any of its coastal relative Mytilus mussel species. Overall, the present study provided new insights into heavy metal and sulfide metabolism in B. platifrons and can be served as the basis for future molecular studies on host-symbiont interactions in cold seep mussels.


Asunto(s)
Inmunidad Innata , Inactivación Metabólica/genética , Metales Pesados/metabolismo , Mytilidae/genética , Sulfuros/metabolismo , Transcriptoma , Secuencia de Aminoácidos , Estructuras Animales/inmunología , Estructuras Animales/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , China , Bases de Datos Genéticas , Ontología de Genes , Branquias/inmunología , Branquias/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Respiraderos Hidrotermales , Metalotioneína/genética , Metalotioneína/metabolismo , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Mytilidae/clasificación , Mytilidae/inmunología , Mytilidae/metabolismo , Océano Pacífico , Peptidoglicano/biosíntesis , Peptidoglicano/aislamiento & purificación , Filogenia , Fitoquelatinas/genética , Fitoquelatinas/metabolismo , Alineación de Secuencia
8.
Retrovirology ; 12: 61, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26169178

RESUMEN

BACKGROUND: Nef is a multifunctional HIV-1 protein critical for progression to AIDS. Humans infected with nef(-) HIV-1 have greatly delayed or no disease consequences. We have contrasted nef(-) and nef(+) infection of BLT humanized mice to better characterize Nef's pathogenic effects. RESULTS: Mice were inoculated with CCR5-tropic HIV-1JRCSF (JRCSF) or JRCSF with an irreversibly inactivated nef (JRCSFNefdd). In peripheral blood (PB), JRCSF exhibited high levels of viral RNA (peak viral loads of 4.71 × 10(6) ± 1.23 × 10(6) copies/ml) and a progressive, 75% loss of CD4(+) T cells over 17 weeks. Similar losses were observed in CD4(+) T cells from bone marrow, spleen, lymph node, lung and liver but thymocytes were not significantly decreased. JRCSFNefdd also had high peak viral loads (2.31 × 10(6) ± 1.67 × 10(6)) but induced no loss of PB CD4(+) T cells. In organs, JRCSFNefdd produced small, but significant, reductions in CD4(+) T cell levels and did not affect the level of thymocytes. Uninfected mice have low levels of HLA-DR(+)CD38(+)CD8(+) T cells in blood (1-2%). Six weeks post inoculation, JRCSF infection resulted in significantly elevated levels of activated CD8(+) T cells (6.37 ± 1.07%). T cell activation coincided with PB CD4(+) T cell loss which suggests a common Nef-dependent mechanism. At 12 weeks, in JRCSF infected animals PB T cell activation sharply increased to 19.7 ± 2.9% then subsided to 5.4 ± 1.4% at 14 weeks. HLA-DR(+)CD38(+)CD8(+) T cell levels in JRCSFNefdd infected mice did not rise above 1-2% despite sustained high levels of viremia. Interestingly, we also noted that in mice engrafted with human tissue expressing a putative protective HLA-B allele (B42:01), JRCSFNefdd exhibited a substantial (200-fold) reduced viral load compared to JRCSF. CONCLUSIONS: Nef expression was necessary for both systemic T cell activation and substantial CD4(+) T cell loss from blood and tissues. JRCSFNefdd infection did not activate CD8(+) T cells or reduce the level of CD4(+) T cells in blood but did result in a small Nef-independent decrease in CD4(+) T cells in organs. These observations strongly support the conclusion that viral pathogenicity is mostly driven by Nef. We also observed for the first time substantial host-specific suppression of HIV-1 replication in a small animal infection model.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Activación de Linfocitos , Replicación Viral , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Estructuras Animales/inmunología , Animales , Sangre/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/virología , Humanos , Ratones , Ratones SCID
9.
J Virol ; 89(11): 5883-94, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25787286

RESUMEN

UNLABELLED: Macrophages regulate tissue immunity, orchestrating the initiation and resolution of antimicrobial immune responses and repair of damaged tissue architecture. Their dysfunction can, thus, manifest in either pro- and anti-inflammatory responses. Indeed, despite the importance of macrophage function in health and disease, the role of tissue-resident macrophages in human immunodeficiency virus (HIV) disease progression remains incompletely defined. Here, we use flow cytometry to assess the phenotypes and functions of macrophages isolated from the spleens, axillary lymph nodes, colons, jejuna, and livers of healthy and chronically simian immunodeficiency virus (SIV)-infected Asian macaques, the prominent nonhuman primate model for HIV infection. Our data demonstrate that macrophages from healthy animals exhibit considerable phenotypic and functional heterogeneity across tissues and across a variety of stimuli. Further, our analysis reveals changes in the lipopolysaccharide (LPS) responsiveness of macrophages isolated from SIV-infected animals. We anticipate that our findings will inform future research into macrophage-directed immunity across a variety of primate diseases. IMPORTANCE: These findings highlight the functional and phenotypic heterogeneity of tissue macrophages in different anatomic sites and as a result of SIV infection. We believe that our data will lead to novel therapeutic interventions aimed at altering the proinflammatory capacity of tissue macrophages in progressively HIV-infected individuals.


Asunto(s)
Macrófagos/clasificación , Macrófagos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Estructuras Animales/inmunología , Animales , Asia , Citometría de Flujo , Inmunofenotipificación , Lipopolisacáridos/inmunología , Macaca mulatta , Macrófagos/química
10.
J Virol ; 88(17): 9490-503, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942579

RESUMEN

UNLABELLED: Virus-specific CD8+ T cells in the lymphoid organs contract at the resolution of virus infections by apoptosis or by dissemination into peripheral tissues, and those residing in nonlymphoid organs, including the peritoneal cavity and fat pads, are more resistant to apoptosis than those in the spleen and lymph nodes. This stability of memory T cells in the nonlymphoid tissues may enhance protection to secondary challenges. Here, we show that lymphocytic choriomeningitis virus (LCMV)-specific CD8+ T cells in nonlymphoid tissues were enriched for memory precursors (expressing high levels of interleukin-7 receptor and low levels of killer cell lectin-like receptor G1 [IL-7Rhi KLRG1lo]) and had higher expression of CD27, CXCR3, and T cell factor-1 (TCF-1), each a marker that is individually correlated with decreased apoptosis. CD8+ T cells in the peritoneal cavity of TCF-1-deficient mice had decreased survival, suggesting a role for TCF-1 in promoting survival in the nonlymphoid tissues. CXCR3+ CD8+ T cells resisted apoptosis and accumulated in the lymph nodes of mice treated with FTY720, which blocks the export of lymph node cells into peripheral tissue. The peritoneal exudate cells (PEC) expressed increased amounts of CXCR3 ligands, CXCL9 and CXCL10, which may normally recruit these nonapoptotic cells from the lymph nodes. In addition, adoptive transfer of splenic CD8+ T cells into PEC or spleen environments showed that the peritoneal environment promoted survival of CD8+ T cells. Thus, intrinsic stability of T cells which are present in the nonlymphoid tissues along with preferential migration of apoptosis-resistant CD8+ T cells into peripheral sites and the availability of tissue-specific factors that enhance memory cell survival may collectively account for the tissue-dependent apoptotic differences. IMPORTANCE: Most infections are initiated at nonlymphoid tissue sites, and the presence of memory T cells in nonlymphoid tissues is critical for protective immunity in various viral infection models. Virus-specific CD8+ T cells in the nonlymphoid tissues are more resistant to apoptosis than those in lymphoid organs during the resolution and memory phase of the immune response to acute LCMV infection. Here, we investigated the mechanisms promoting stability of T cells in the nonlymphoid tissues. This increased resistance to apoptosis of virus-specific CD8+ T cells in nonlymphoid tissues was due to several factors. Nonlymphoid tissues were enriched in memory phenotype CD8+ T cells, which were intrinsically resistant to apoptosis irrespective of the tissue environment. Furthermore, apoptosis-resistant CD8+ T cells preferentially migrated into the nonlymphoid tissues, where the availability of tissue-specific factors may enhance memory cell survival. Our findings are relevant for the generation of long-lasting vaccines providing protection at peripheral infection sites.


Asunto(s)
Estructuras Animales/inmunología , Apoptosis , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/fisiología , Virus de la Coriomeningitis Linfocítica/inmunología , Subgrupos de Linfocitos T/fisiología , Estructuras Animales/patología , Animales , Antígenos de Superficie/análisis , Infecciones por Arenaviridae/patología , Linfocitos T CD8-positivos/química , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Masculino , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología
11.
Infect Immun ; 82(8): 3275-88, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24866796

RESUMEN

The signaling mechanisms regulating neutrophil recruitment, systemic inflammation, and T-cell dysfunction in polymicrobial sepsis are not clear. This study explored the potential involvement of the calcium/calcineurin-dependent transcription factor, nuclear factor of activated T cells (NFAT), in abdominal sepsis. Cecal ligation and puncture (CLP) triggered NFAT-dependent transcriptional activity in the lung, spleen, liver, and aorta in NFAT-luciferase reporter mice. Treatment with the NFAT inhibitor A-285222 prior to CLP completely prevented sepsis-induced NFAT activation in all these organs. Inhibition of NFAT activity reduced sepsis-induced formation of CXCL1, CXCL2, and CXCL5 chemokines and edema as well as neutrophil infiltration in the lung. Notably, NFAT inhibition efficiently reduced the CLP-evoked increases in HMBG1, interleukin 6 (IL-6), and CXCL5 levels in plasma. Moreover, administration of A-285222 restored sepsis-induced T-cell dysfunction, as evidenced by markedly decreased apoptosis and restored proliferative capacity of CD4 T cells. Along these lines, treatment with A-285222 restored gamma interferon (IFN-γ) and IL-4 levels in the spleen, which were markedly reduced in septic mice. CLP-induced formation of regulatory T cells (CD4(+) CD25(+) Foxp3(+)) in the spleen was also abolished in A-285222-treated animals. All together, these novel findings suggest that NFAT is a powerful regulator of pathological inflammation and T-cell immune dysfunction in abdominal sepsis. Thus, our data suggest that NFAT signaling might be a useful target to protect against respiratory failure and immunosuppression in patients with sepsis.


Asunto(s)
Factores de Transcripción NFATC/metabolismo , Infiltración Neutrófila , Peritonitis/inmunología , Peritonitis/patología , Sepsis/inmunología , Sepsis/patología , Linfocitos T/inmunología , Estructuras Animales/inmunología , Estructuras Animales/patología , Animales , Coinfección/inmunología , Coinfección/patología , Inflamación , Ratones
12.
Virol Sin ; 29(1): 48-60, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24452537

RESUMEN

The suppressive capacity of regulatory T cells (Tregs) has been extensively studied and is well established for many diseases. The expansion, accumulation, and activation of Tregs in viral infections are of major interest in order to find ways to alter Treg functions for therapeutic benefit. Tregs are able to dampen effector T cell responses to viral infections and thereby contribute to the establishment of a chronic infection. In the Friend retrovirus (FV) mouse model, Tregs are known to expand in all infected organs. To better understand the characteristics of these Treg populations, their phenotype was analyzed in detail. During acute FV-infection, Tregs became activated in the spleen and bone marrow, as indicated by various T cell activation markers, such as CD43 and CD103. Interestingly, Tregs in the bone marrow, which contains the highest viral loads during acute infection, displayed greater levels of activation than Tregs from the spleen. Treg expansion was driven by proliferation but no FV-specific Tregs could be detected. Activated Tregs in FV-infection did not produce Granzyme B (GzmB) or tumor necrosis factor α (TNFα), which are thought to be a potential mechanism for their suppressive activity. Furthermore, Tregs expressed inhibitory markers, such as TIM3, PD-1 and PD-L1. Blocking TIM3 and PD-L1 with antibodies during chronic FV-infection increased the numbers of activated Tregs. These data may have important implications for the understanding of Treg functions during chronic viral infections.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Leucemia Experimental/inmunología , Activación de Linfocitos , Infecciones por Retroviridae/inmunología , Linfocitos T Reguladores/inmunología , Infecciones Tumorales por Virus/inmunología , Estructuras Animales/inmunología , Estructuras Animales/virología , Animales , Antígeno B7-H1/análisis , Médula Ósea/inmunología , Médula Ósea/virología , Granzimas/análisis , Receptor 2 Celular del Virus de la Hepatitis A , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/análisis , Receptores Virales/análisis , Bazo/inmunología , Bazo/virología , Linfocitos T Reguladores/química , Factor de Necrosis Tumoral alfa/análisis
13.
Cell Microbiol ; 16(5): 687-700, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24003897

RESUMEN

Infection with the malaria parasite, Plasmodium, is associated with a strong inflammatory response and parasite cytoadhesion (sequestration) in several organs. Here, we have carried out a systematic study of sequestration and histopathology during infection of C57Bl/6 mice with Plasmodium chabaudi AS and determined the influence of the immune response. This parasite sequesters predominantly in liver and lung, but not in the brain, kidney or gut. Histopathological changes occur in multiple organs during the acute infection, but are not restricted to the organs where sequestration takes place. Adaptive immunity, and signalling through the IFNγ receptor increased sequestration and histopathology in the liver, but not in the lung, suggesting that there are differences in the adhesion molecules and/or parasite ligands utilized and mechanisms of pathogenesis in these two organs. Exacerbation of pro-inflammatory responses during infection by deletion of the il10 gene resultsin the aggravation of damage to lung and kidney irrespective of the degree of sequestration. The immune response therefore affected both sequestration and histopathology in an organ-specific manner. P. chabaudi AS provides a good model to investigate the influence of the host response on the sequestration and specific organ pathology, which is applicable to human malaria.


Asunto(s)
Estructuras Animales/inmunología , Malaria/inmunología , Malaria/patología , Plasmodium chabaudi/inmunología , Estructuras Animales/parasitología , Estructuras Animales/patología , Animales , Histocitoquímica , Ratones , Ratones Endogámicos C57BL
14.
Mar Biotechnol (NY) ; 16(1): 10-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23900602

RESUMEN

The brightness and color of pearls varies among different pearl-producing shellfish and have been a source of human fascination since ancient times. When produced through cultivation, the characteristics and quality of a pearl depend on the kind of shellfish used and also the transplanted mantle graft. This suggests that the Akoya pearl oyster, which is generally used in Japan for pearl culturing, can produce different kinds of pearl through the use of mantles from different species of shellfish. However, a transplanted heterogeneous mantle would be rejected by the immune system of the Akoya oyster. We have therefore developed a new method to suppress the Akoya immune system that archives immune tolerance to other shellfish. It is generally known that small quantities of antigens can be used to produce archived immunological tolerance in a clinical setting. We successfully suppressed the Akoya pearl oyster immune response against a Mabé pearl oyster graft through repeat injections of mantle homogenates. We then transplanted a Mabé pearl oyster mantle graft into the immunologically tolerant Akoya pearl oyster and obtained a Mabé pearl from an Akoya pearl oyster. Our new technique thus makes the production of novel and different pearls in the Akoya possible. We believe that this has significant future potential for the advancement of the pearl industry.


Asunto(s)
Exoesqueleto/química , Estructuras Animales/trasplante , Acuicultura/métodos , Pinctada/química , Pinctada/inmunología , Estructuras Animales/inmunología , Animales , Cartilla de ADN/genética , Citometría de Flujo , Fluorescencia , Terapia de Inmunosupresión/métodos , Fagocitosis/fisiología , Reacción en Cadena de la Polimerasa , Especificidad de la Especie , Trasplante Heterólogo
15.
Infect Immun ; 81(9): 3173-81, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23774598

RESUMEN

Sepsis is an infection-induced systemic inflammatory response syndrome. Upstream recognition molecules, like CD14, play key roles in the pathogenesis. The aim of the present study was to investigate the effect of systemic CD14 inhibition on local inflammatory responses in organs from septic pigs. Pigs (n = 34) receiving Escherichia coli-bacteria or E. coli-lipopolysaccharide (LPS) were treated with an anti-CD14 monoclonal antibody or an isotype-matched control. Lungs, liver, spleen, and kidneys were examined for bacteria and inflammatory biomarkers. E. coli and LPS were found in large amounts in the lungs compared to the liver, spleen, and kidneys. Notably, the bacterial load did not predict the respective organ inflammatory response. There was a marked variation in biomarker induction in the organs and in the effect of anti-CD14. Generally, the spleen produced the most cytokines per weight unit, whereas the liver contributed the most to the total load. All cytokines were significantly inhibited in the spleen. Interleukin-6 (IL-6) was significantly inhibited in all organs, IL-1ß and IP-10 were significantly inhibited in liver, spleen, and kidneys, and tumor necrosis factor, IL-8, and PAI-1 were inhibited only in the spleen. ICAM-1 and VCAM-1 was significantly inhibited in the kidneys. Systemic CD14-inhibition efficiently, though organ dependent, attenuated local inflammatory responses. Detailed knowledge on how the different organs respond to systemic inflammation in vivo, beyond the information gained by blood examination, is important for our understanding of the nature of systemic inflammation and is required for future mediator-directed therapy in sepsis. Inhibition of CD14 seems to be a good candidate for such treatment.


Asunto(s)
Escherichia coli/inmunología , Inflamación/inmunología , Receptores de Lipopolisacáridos/inmunología , Sepsis/inmunología , Porcinos/inmunología , Estructuras Animales/inmunología , Estructuras Animales/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Biomarcadores/metabolismo , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Escherichia coli/metabolismo , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucinas/inmunología , Interleucinas/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Inhibidor 1 de Activador Plasminogénico/inmunología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sepsis/metabolismo , Porcinos/metabolismo , Molécula 1 de Adhesión Celular Vascular/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Dev Comp Immunol ; 41(1): 59-67, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23562573

RESUMEN

Peroxinectins function as hemoperoxidase and cell adhesion factor involved in invertebrate immune reaction. In this study, the ascidian (Ciona intestinalis) peroxinectin gene (CiPxt) and its expression during the inflammatory response have been examined. CiPxt is a new member of the peroxidase-cyclooxygenase gene superfamily that contains both the peroxidase domain and the integrin KGD (Lys-Gly-Asp) binding motif. A phylogenetic tree showed that CiPxt is very close to the chordate group and appears to be the outgroup of mammalian MPO, EPO and TPO clades. The CiPxt molecular structure model resulted superimposable to the human myeloperoxidase. The CiPxt mRNA expression is upregulated by LPS inoculation suggesting it is involved in C. intestinalis inflammatory response. The CiPxt was expressed in hemocytes (compartment/morula cells), vessel epithelium, and unilocular refractile granulocytes populating the inflamed tunic matrix and in the zones 7, 8 and 9 of the endostyle, a special pharynx organs homolog to the vertebrate thyroid gland.


Asunto(s)
Estructuras Animales/inmunología , Moléculas de Adhesión Celular/inmunología , Ciona intestinalis/inmunología , Hemocitos/inmunología , Peroxidasas/inmunología , ARN Mensajero/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Estructuras Animales/efectos de los fármacos , Animales , Moléculas de Adhesión Celular/clasificación , Moléculas de Adhesión Celular/genética , Ciona intestinalis/anatomía & histología , Ciona intestinalis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Hemocitos/citología , Hemocitos/efectos de los fármacos , Inmunidad Innata , Lipopolisacáridos/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Especificidad de Órganos , Peroxidasas/clasificación , Peroxidasas/genética , Filogenia , Estructura Terciaria de Proteína , ARN Mensajero/genética , Homología de Secuencia de Aminoácido
17.
Microb Pathog ; 59-60: 33-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23583292

RESUMEN

Understanding of uptake and invasion routes of Yersinia ruckeri, causing Enteric Red Mouth Disease (ERM) in rainbow trout (Oncorhynchus mykiss), is essential for improved understanding of the pathogenicity and immune response mechanisms associated this disease. The present work shed light on areas of invasion in rainbow trout by the use of immunohistochemistry and in situ hybridization techniques. Fish were exposed to live or formalin inactivated bacteria and samples were subsequently taken for histology from various outer and inner surfaces. We applied a specific monoclonal antibody and specific oligonucleotide probes binding to Y. ruckeri (serotype O1, biotype 2) in tissue sections and were able to demonstrate a tissue specific uptake of this bacterium (both formalin inactivated and live form). Uptake and subsequent translocation dynamics at various surfaces demonstrated different site specific propensities between the formalin inactivated and live bacterial organisms. Lateral lines, dorsal fin, epidermis and gastro-intestinal tract mucosal tissue were the primary areas where bacterial uptake was demonstrated readily after exposure. The fate of internalized bacterial organisms within the host suggested that central immune organs are involved in the final antigen processing.


Asunto(s)
Estructuras Animales/microbiología , Enfermedades de los Peces/microbiología , Oncorhynchus mykiss/microbiología , Yersiniosis/microbiología , Yersinia ruckeri/aislamiento & purificación , Yersinia ruckeri/patogenicidad , Estructuras Animales/inmunología , Animales , Modelos Animales de Enfermedad , Enfermedades de los Peces/inmunología , Inmunohistoquímica , Hibridación in Situ , Oncorhynchus mykiss/inmunología , Yersiniosis/inmunología , Yersinia ruckeri/inmunología
18.
BMC Infect Dis ; 13: 147, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23521724

RESUMEN

BACKGROUND: Candidemia is a severe fungal infection that primarily affects hospitalized and/or immunocompromised patients. Mononuclear phagocytes have been recognized as pivotal immune cells which act in the recognition of pathogens, phagocytosis, inflammation, polarization of adaptive immune response and tissue repair. Experimental studies have showed that the systemic candidiasis could be controlled by activated peritoneal macrophages. However, the mechanism to explain how these cells act in distant tissue during a systemic fungal infection is still to be elucidated. In the present study we investigate the in vivo trafficking of phagocytic peritoneal cells into infected organs in hypoinsulinemic-hyperglycemic (HH) mice with systemic candidiasis. METHODS: The red fluorescent vital dye PKH-26 PCL was injected into the peritoneal cavity of Swiss mice 24 hours before the intravenous inoculation with Candida albicans. After 24 and 48 hours and 7 days of infection, samples of the spleen, liver, kidneys, brain and lungs were submitted to the microbiological evaluation as well as to phagocytic peritoneal cell trafficking analyses by fluorescence microscopy. RESULTS: In the present study, PKH+ cells were observed in the peritoneum, kidney, spleen and liver samples from all groups. In infected mice, we also found PKH+ cells in the lung and brain. The HH condition did not affect this process. CONCLUSIONS: In the present study we have observed that peritoneal phagocytes migrate to tissues infected by C. albicans and the HH condition did not interfere in this process.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Macrófagos Peritoneales/inmunología , Estructuras Animales/inmunología , Estructuras Animales/microbiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos
19.
Innate Immun ; 19(6): 569-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23405032

RESUMEN

Purple sea urchins (Strongylocentrotus purpuratus) express a highly variable set of immune genes called Sp185/333 by two subtypes of coelomocytes: the polygonal and small phagocytes. We report that the Sp185/333 genes and their encoded proteins are also expressed in all of the major organs in the adult sea urchin, including the axial organ, pharynx, esophagus, intestine and gonads. After immune challenge, there is an increase in the level of Sp185/333 mRNA in cells associated with the intestine and axial organ. The Sp185/333 proteins increase in the axial organ, pharynx, esophagus and intestine after challenge. However, the proportion of Sp185/333-positive cells only increases in the axial organ, while there is no change in that proportion in the other organs after challenge. The size range of the major Sp185/333 proteins expressed by organs is broader (5 kDa to > 250 kDa) compared with those in coelomocytes (∼40 kDa to < 250 kDa). Images of the different organs do not clarify whether coelomocytes or parenchymal cells express the Sp185/333 proteins. The increase in levels of Sp185/333 transcripts, protein expression and Sp185/333-positive cells in the axial organ in response to challenge suggests that this organ may have an important role in immunity for this species.


Asunto(s)
Estructuras Animales/inmunología , Sistema Inmunológico/fisiología , Strongylocentrotus purpuratus/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Variación Genética , Inmunidad Innata/genética , Lipopolisacáridos/inmunología , Transcriptoma
20.
Microbiol Immunol ; 56(8): 562-71, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22671916

RESUMEN

Viral diseases restrict the development of the world shrimp industry and there are few studies on cell response to the presence of viral infections. We performed immunohistochemistry assays to characterize hemocytes subpopulations involved in the immune process occurring in the LO of Litopenaeus vannamei shrimp. Tissue sections of animals that increased their LO spheroids and hemocytes infiltration after WSSV induced infection, were used. Three MABs namely, 40E10 (recognizing small granule hemocytes), 40E2 (recognizing large granule hemocytes), and 41B12, which recognize α(2)-macroglobulin were used. Additionally one polyclonal antibody was used against the penaeidins antimicrobial peptides, and to detect WSSV a commercial immunohistochemistry kit (DiagXotics) was used. Numerous small granule hemocytes were detected in the stromal matrix of LO tubules, whereas large granule hemocytes were less numerous and located mainly in hemal sinuses. The exocytosis of two molecules, which have been related to the phagocytosis process, i.e. penaeidins, and α(2)-macroglobulin, was detected in the external stromal matrix and the outer tubule walls. α(2) -macroglobulin inhibits phenoloxidase activity and its strong release in LO tissue may explain the absence of melanization in the immune processes occurring in it. The immunolabeling of vesicles within the LO spheroids with MABs 41B12 40E10 and antipenaedin antibody suggests that LOS are formed by phagocytic cells derived from small granule and hyaline hemocytes, with a possible role of peneidins and α(2)-macroglobulin acting as opsonines.


Asunto(s)
Hemocitos/citología , Penaeidae/virología , Virus del Síndrome de la Mancha Blanca 1/inmunología , alfa-Macroglobulinas/análisis , Estructuras Animales/citología , Estructuras Animales/inmunología , Animales , Anticuerpos Monoclonales , Inmunohistoquímica , Microscopía , Penaeidae/inmunología , Coloración y Etiquetado/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...