Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38612744

RESUMEN

The mission of this review is to identify immune-damaging participants involved in antiviral immunoinflammatory lesions. We argue these could be targeted and their activity changed selectively by maneuvers that, at the same time, may not diminish the impact of components that help resolve lesions. Ideally, we need to identify therapeutic approaches that can reverse ongoing lesions that lack unwanted side effects and are affordable to use. By understanding the delicate balance between immune responses that cause tissue damage and those that aid in resolution, novel strategies can be developed to target detrimental immune components while preserving the beneficial ones. Some strategies involve rebalancing the participation of immune components using various approaches, such as removing or blocking proinflammatory T cell products, expanding regulatory cells, restoring lost protective cell function, using monoclonal antibodies (moAb) to counteract inhibitory molecules, and exploiting metabolic differences between inflammatory and immuno-protective responses. These strategies can help reverse ongoing viral infections. We explain various approaches, from model studies and some clinical evidence, that achieve innate and adaptive immune rebalancing, offering insights into potential applications for controlling chronic viral-induced lesions.


Asunto(s)
Anticuerpos Monoclonales , Pirimetamina , Humanos , Anticuerpos Monoclonales/uso terapéutico , Sulfadiazina
2.
Front Immunol ; 14: 1257192, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37671156

RESUMEN

In this review, we discuss a variety of immune modulating approaches that could be used to counteract tissue-damaging viral immunoinflammatory lesions which typify many chronic viral infections. We make the point that in several viral infections the lesions can be largely the result of one or more aspects of the host response mediating the cell and tissue damage rather than the virus itself being directly responsible. However, within the reactive inflammatory lesions along with the pro-inflammatory participants there are also other aspects of the host response that may be acting to constrain the activity of the damaging components and are contributing to resolution. This scenario should provide the prospect of rebalancing the contributions of different host responses and hence diminish or even fully control the virus-induced lesions. We identify several aspects of the host reactions that influence the pattern of immune responsiveness and describe approaches that have been used successfully, mainly in model systems, to modulate the activity of damaging participants and which has led to lesion control. We emphasize examples where such therapies are, or could be, translated for practical use in the clinic to control inflammatory lesions caused by viral infections.


Asunto(s)
Modelos Biológicos , Pirimetamina , Humanos , Sulfadiazina
4.
Int J Mol Sci ; 23(21)2022 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-36361628

RESUMEN

It is now widely accepted that NK cells can acquire memory, and this makes them more effective to protect against some pathogens. Prior reports indicate memory-like NK cells (mlNKs) in murine model of Mycobacterium tuberculosis (Mtb) as well as in healthy individuals with latent TB infection (LTBI). The increased expression of CD226 was evident in mlNKs from LTBI+ people after stimulation with γ-irradiated Mtb (γ-Mtb). We thus evaluated the contribution of costimulatory CD226 signaling in the functionality of mlNKs in LTBI+ people. We found that blockade of CD226 signaling using the antibody- or CRISPR/Cas9-mediated deletion of the CD226 gene in NK cells diminished the proliferation of mlNKs from LTBI+ people. Blocking CD226 signaling also reduced the phosphorylation of FOXO1 and cMyc expression. Additionally, cMyc inhibition using a chemical inhibitor reduced proliferation by mlNKs from LTBI+ people. Moreover, blocking CD226 signaling reduced glycolysis in NK cells, and the inhibition of glycolysis led to reduced effector function of mlNKs from LTBI+ people. Overall, our results provide a role for CD226 signaling in mlNK responses to Mtb.


Asunto(s)
Tuberculosis Latente , Mycobacterium tuberculosis , Humanos , Ratones , Animales , Tuberculosis Latente/microbiología , Células Asesinas Naturales , Transducción de Señal , Proliferación Celular
6.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34785597

RESUMEN

Zika virus (ZIKV) during pregnancy infects fetal trophoblasts and causes placental damage and birth defects including microcephaly. Little is known about the anti-ZIKV cellular immune response at the maternal-fetal interface. Decidual natural killer cells (dNK), which directly contact fetal trophoblasts, are the dominant maternal immune cells in the first-trimester placenta, when ZIKV infection is most hazardous. Although dNK express all the cytolytic molecules needed to kill, they usually do not kill infected fetal cells but promote placentation. Here, we show that dNK degranulate and kill ZIKV-infected placental trophoblasts. ZIKV infection of trophoblasts causes endoplasmic reticulum (ER) stress, which makes them dNK targets by down-regulating HLA-C/G, natural killer (NK) inhibitory receptor ligands that help maintain tolerance of the semiallogeneic fetus. ER stress also activates the NK activating receptor NKp46. ZIKV infection of Ifnar1 -/- pregnant mice results in high viral titers and severe intrauterine growth restriction, which are exacerbated by depletion of NK or CD8 T cells, indicating that killer lymphocytes, on balance, protect the fetus from ZIKV by eliminating infected cells and reducing the spread of infection.


Asunto(s)
Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Feto/inmunología , Antígenos HLA-C , Tolerancia Inmunológica , Ratones , Placenta/inmunología , Placentación , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Receptores KIR
7.
Am J Respir Cell Mol Biol ; 64(4): 477-491, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33600743

RESUMEN

Streptococcus pneumoniae is the leading cause of hospital community-acquired pneumonia. Patients with pneumococcal pneumonia may develop complicated parapneumonic effusions or empyema that can lead to pleural organization and subsequent fibrosis. The pathogenesis of pleural organization and scarification involves complex interactions between the components of the immune system, coagulation, and fibrinolysis. EPCR (endothelial protein C receptor) is a critical component of the protein C anticoagulant pathway. The present study was performed to evaluate the role of EPCR in the pathogenesis of S. pneumoniae infection-induced pleural thickening and fibrosis. Our studies show that the pleural mesothelium expresses EPCR. Intrapleural instillation of S. pneumoniae impairs lung compliance and lung volume in wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. Intrapleural S. pneumoniae infection induces pleural thickening in wild-type mice. Pleural thickening is more pronounced in EPCR-overexpressing mice, whereas it is reduced in EPCR-deficient mice. Markers of mesomesenchymal transition are increased in the visceral pleura of S. pneumoniae-infected wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. The lungs of wild-type and EPCR-overexpressing mice administered intrapleural S. pneumoniae showed increased infiltration of macrophages and neutrophils, which was significantly reduced in EPCR-deficient mice. An analysis of bacterial burden in the pleural lavage, the lungs, and blood revealed a significantly lower bacterial burden in EPCR-deficient mice compared with wild-type and EPCR-overexpressing mice. Overall, our data provide strong evidence that EPCR deficiency protects against S. pneumoniae infection-induced impairment of lung function and pleural remodeling.


Asunto(s)
Receptor de Proteína C Endotelial/deficiencia , Pulmón/metabolismo , Pleura/metabolismo , Derrame Pleural/metabolismo , Pleuresia/metabolismo , Neumonía Neumocócica/metabolismo , Streptococcus pneumoniae/patogenicidad , Animales , Carga Bacteriana , Células Cultivadas , Modelos Animales de Enfermedad , Receptor de Proteína C Endotelial/genética , Femenino , Fibrosis , Interacciones Huésped-Patógeno , Humanos , Pulmón/microbiología , Pulmón/patología , Pulmón/fisiopatología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/microbiología , Pleura/microbiología , Pleura/patología , Derrame Pleural/microbiología , Derrame Pleural/patología , Derrame Pleural/fisiopatología , Pleuresia/microbiología , Pleuresia/patología , Pleuresia/fisiopatología , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/patología , Neumonía Neumocócica/fisiopatología
8.
PLoS Pathog ; 16(12): e1009132, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370392

RESUMEN

NK cells have been shown to display adaptive traits such as memory formation akin to T and B lymphocytes. Here we show that Zika virus infection induces memory like NK cells that express CD27. Strikingly, these cells exhibit stem-like features that include expansion capacity, self-renewal pathway, differentiation into effector cells, longer telomeres and gene signature associated with hematopoietic stem cell (HSC) progenitors. This subset shared transcriptional and epigenetic changes with memory CD8 T cells, stem cells and stem like T cells. These NK cells with memory and stem cell features, which we term "NK memory stem cells", demonstrated greater antiviral potential than CD27- or naïve CD27+ NK when adoptively transferred to Zika infected mice. Our results also suggest a role for the transcription factor TCF-1 in memory and stemness features of this NK subset. This study defines a unique TCF1hi CD27+ NK subset with memory capacity and stem cell features that play a role in antiviral immunity.


Asunto(s)
Memoria Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Células Madre/inmunología , Infección por el Virus Zika/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
9.
Am J Physiol Lung Cell Mol Physiol ; 319(6): L893-L907, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32996778

RESUMEN

Inhalation of organic dust is an occupational hazard leading to the development of respiratory symptoms and respiratory diseases. Bioaerosols from concentrated animal feeding operations are rich in bacteria and could carry bacterial extracellular vesicles (EVs) that could induce lung inflammation. It is not known if organic dust contains bacterial EVs and whether they modulate lung inflammation. Herein, we show that poultry organic dust contains bacterial EVs (dust EVs) that induce lung inflammation. Treatment of airway epithelial cells, THP-1-monocytes and -macrophages with dust EVs rapidly induced IL-8, IL-6, ICAM-1, proIL-1ß, and TNF-α levels. In airway epithelial cells, induction of inflammatory mediators was due to increased mRNA levels and NF-κB activation. Induction of inflammatory mediators by dust EVs was not inhibited by polymyxin B. Single and repeated treatments of mice with dust EVs increased lung KC, IL-6, and TNF-α levels without significantly altering IL-17A levels. Increases in cytokines were associated with enhanced neutrophil infiltration into the lung. Repeated treatments of mice with dust EVs increased lung mean linear intercept and increased collagen deposition around airways indicating lung remodeling. Peribronchial cell infiltrates and airway epithelial thickening were also observed in treated mice. Because bacterial EVs are nanometer-sized particles, they can reach and accumulate in the bronchiolar and alveolar regions causing lung injury leading to the development of respiratory diseases. Our studies have provided new evidence for the presence of bacterial EVs in organic dust and for their role as one of the causative agents of organic dust-induced lung inflammation and lung injury.


Asunto(s)
Citocinas/metabolismo , Inflamación/metabolismo , Pulmón/metabolismo , Neumonía/metabolismo , Animales , Células Epiteliales/metabolismo , Mediadores de Inflamación/farmacología , Macrófagos/metabolismo , Ratones , Monocitos/metabolismo , Neutrófilos/metabolismo , Neumonía/inducido químicamente
10.
Cell ; 182(5): 1125-1139.e18, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32822574

RESUMEN

Maternal decidual NK (dNK) cells promote placentation, but how they protect against placental infection while maintaining fetal tolerance is unclear. Here we show that human dNK cells highly express the antimicrobial peptide granulysin (GNLY) and selectively transfer it via nanotubes to extravillous trophoblasts to kill intracellular Listeria monocytogenes (Lm) without killing the trophoblast. Transfer of GNLY, but not other cell death-inducing cytotoxic granule proteins, strongly inhibits Lm in human placental cultures and in mouse and human trophoblast cell lines. Placental and fetal Lm loads are lower and pregnancy success is greatly improved in pregnant Lm-infected GNLY-transgenic mice than in wild-type mice that lack GNLY. This immune defense is not restricted to pregnancy; peripheral NK (pNK) cells also transfer GNLY to kill bacteria in macrophages and dendritic cells without killing the host cell. Nanotube transfer of GNLY allows dNK to protect against infection while leaving the maternal-fetal barrier intact.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Bacterias/inmunología , Movimiento Celular/inmunología , Células Asesinas Naturales/inmunología , Trofoblastos/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Células HeLa , Humanos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Placenta/inmunología , Placenta/microbiología , Embarazo , Ratas , Células THP-1 , Trofoblastos/microbiología
11.
Nat Commun ; 11(1): 3535, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32669568

RESUMEN

Macrophages are professional phagocytes known to play a vital role in controlling Mycobacterium tuberculosis (Mtb) infection and disease progression. Here we compare Mtb growth in mouse alveolar (AMs), peritoneal (PMs), and liver (Kupffer cells; KCs) macrophages and in bone marrow-derived monocytes (BDMs). KCs restrict Mtb growth more efficiently than all other macrophages and monocytes despite equivalent infections through enhanced autophagy. A metabolomics comparison of Mtb-infected macrophages indicates that ornithine and imidazole are two top-scoring metabolites in Mtb-infected KCs and that acetylcholine is the top-scoring in Mtb-infected AMs. Ornithine, imidazole and atropine (acetylcholine inhibitor) inhibit Mtb growth in AMs. Ornithine enhances AMPK mediated autophagy whereas imidazole directly kills Mtb by reducing cytochrome P450 activity. Intranasal delivery of ornithine or imidazole or the two together restricts Mtb growth. Our study demonstrates that the metabolic differences between Mtb-infected AMs and KCs lead to differences in the restriction of Mtb growth.


Asunto(s)
Autofagia/efectos de los fármacos , Ornitina/farmacología , Tuberculosis/tratamiento farmacológico , Urea/química , Amoníaco/química , Animales , Apoptosis , Arginasa/química , Atropina/farmacología , Proliferación Celular , Progresión de la Enfermedad , Femenino , Imidazoles/farmacología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/microbiología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/microbiología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/microbiología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/química , Fosfatidilserinas/química , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/química
12.
JCI Insight ; 5(5)2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32161191

RESUMEN

Diabetes is a significant risk factor for the development of active tuberculosis. In this study, we used a mouse model of type 2 diabetes mellitus (T2DM) to determine the effect of prior Bacillus Calmette-Guérin (BCG) vaccination on immune responses to Mycobacterium tuberculosis (Mtb) infection. We found that, at 6-7 months after Mtb infection, 90% of the Mtb-infected T2DM mice died, whereas only 50% of BCG-vaccinated T2DM-Mtb-infected mice died. Moreover, 40% of the PBS-treated uninfected T2DM mice and 30% of the uninfected BCG-vaccinated T2DM mice died, whereas all uninfected and infected nondiabetic mice survived. BCG vaccination was less effective in reducing the lung bacterial burden of Mtb-infected T2DM mice compared with Mtb-infected nondiabetic mice. BCG vaccination significantly reduced lung inflammation in Mtb-infected T2DM mice compared with that of unvaccinated T2DM mice infected with Mtb. Furthermore, reduced mortality of BCG-vaccinated Mtb-infected T2DM mice is associated with expansion of IL-13-producing CXCR3+ Tregs in the lungs of Mtb-infected T2DM mice. Recombinant IL-13 and Tregs from BCG-vaccinated Mtb-infected T2DM mice converted proinflammatory M1 macrophages to antiinflammatory M2 macrophages. Our findings suggest a potentially novel role for BCG in preventing excess inflammation and mortality in T2DM mice infected with Mtb.


Asunto(s)
Vacuna BCG/administración & dosificación , Diabetes Mellitus Tipo 2/complicaciones , Tuberculosis/mortalidad , Animales , Vacuna BCG/inmunología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Tuberculosis/complicaciones , Tuberculosis/inmunología , Tuberculosis/prevención & control
13.
Cell Rep ; 15(8): 1715-27, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27184854

RESUMEN

Despite their antiviral effect, the in vivo effect of interferons on HIV transmission is difficult to predict, because interferons also activate and recruit HIV-susceptible cells to sites of infection. HIV does not normally induce type I interferons in infected cells, but does if TREX1 is knocked down. Here, we investigated the effect of topical TREX1 knockdown and local interferon production on HIV transmission in human cervicovaginal explants and humanized mice. In explants in which TREX1 was knocked down, HIV induced interferons, which blocked infection. In humanized mice, even though TREX1 knockdown increased infiltrating immune cells, it delayed viral replication for 3-4 weeks. Similarly intravaginal application of type I interferons the day before HIV infection induced interferon responsive genes, reduced inflammation, and decreased viral replication. However, intravenous interferon enhanced inflammation and infection. Thus, in models of human sexual transmission, a localized interferon response inhibits HIV transmission but systemic interferons do not.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , Técnicas de Silenciamiento del Gen , Infecciones por VIH/enzimología , Infecciones por VIH/virología , Interferón beta/metabolismo , Fosfoproteínas/metabolismo , Animales , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Cuello del Útero/patología , Quimera , Femenino , Regulación de la Expresión Génica , VIH/fisiología , Infecciones por VIH/patología , Infecciones por VIH/transmisión , Humanos , Interferón beta/genética , Macrófagos/metabolismo , Ratones , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Vagina/patología , Replicación Viral
14.
Nat Med ; 22(2): 210-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26752517

RESUMEN

Protozoan infections are a serious global health problem. Natural killer (NK) cells and cytolytic T lymphocytes (CTLs) eliminate pathogen-infected cells by releasing cytolytic granule contents--granzyme (Gzm) proteases and the pore-forming perforin (PFN)--into the infected cell. However, these cytotoxic molecules do not kill intracellular parasites. CD8(+) CTLs protect against parasite infections in mice primarily by secreting interferon (IFN)-γ. However, human, but not rodent, cytotoxic granules contain the antimicrobial peptide granulysin (GNLY), which selectively destroys cholesterol-poor microbial membranes, and GNLY, PFN and Gzms rapidly kill intracellular bacteria. Here we show that GNLY delivers Gzms into three protozoan parasites (Trypanosoma cruzi, Toxoplasma gondii and Leishmania major), in which the Gzms generate superoxide and inactivate oxidative defense enzymes to kill the parasite. PFN delivers GNLY and Gzms into infected cells, and GNLY then delivers Gzms to the intracellular parasites. Killer cell-mediated parasite death, which we term 'microbe-programmed cell death' or 'microptosis', is caspase independent but resembles mammalian apoptosis, causing mitochondrial swelling, transmembrane potential dissipation, membrane blebbing, phosphatidylserine exposure, DNA damage and chromatin condensation. GNLY-transgenic mice are protected against infection by T. cruzi and T. gondii, and survive infections that are lethal to wild-type mice. Thus, GNLY-, PFN- and Gzm-mediated elimination of intracellular protozoan parasites is an unappreciated immune defense mechanism.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Granzimas/inmunología , Células Asesinas Naturales/inmunología , Leishmania major , Perforina/inmunología , Linfocitos T Citotóxicos/inmunología , Toxoplasma , Trypanosoma cruzi , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Enfermedad de Chagas/inmunología , Humanos , Leishmaniasis Cutánea/inmunología , Ratones , Ratones Transgénicos , Toxoplasmosis/inmunología
15.
Am J Pathol ; 185(4): 1073-84, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25700796

RESUMEN

Ocular infection with herpes simplex virus 1 can result in a chronic immunoinflammatory stromal keratitis (SK) lesion that is a significant cause of human blindness. A key to controlling SK lesion severity is to identify cellular and molecular events responsible for tissue damage and to manipulate them therapeutically. Potential targets for therapy are miRNAs, but these are minimally explored especially in responses to infection. Here, we demonstrated that Mir155 expression was up-regulated after ocular herpes simplex virus 1 infection, with the increased Mir155 expression occurring mainly in macrophages and CD4(+) T cells and to a lesser extent in neutrophils. In vivo studies indicated that Mir155 knockout mice were more resistant to herpes SK with marked suppression of T helper cells type 1 and 17 responses both in the ocular lesions and the lymphoid organs. The reduced SK lesion severity was reflected by increased phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase 1 and interferon-γ receptor α-chain levels in activated CD4(+) T cells in the lymph nodes. Finally, in vivo silencing of miR-155 by the provision of antagomir-155 nanoparticles to herpes simplex virus 1-infected mice led to diminished SK lesions and corneal vascularization. In conclusion, our results indicate that miR-155 contributes to the pathogenesis of SK and represents a promising target to control SK severity.


Asunto(s)
Sustancia Propia/patología , Sustancia Propia/virología , Queratitis Herpética/genética , Queratitis Herpética/virología , MicroARNs/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Quimiocinas/metabolismo , Sustancia Propia/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Herpesvirus Humano 1/fisiología , Humanos , Inflamación/patología , Inositol Polifosfato 5-Fosfatasas , Queratitis Herpética/inmunología , Queratitis Herpética/patología , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Modelos Biológicos , Nanopartículas/química , Oligonucleótidos/farmacología , Monoéster Fosfórico Hidrolasas/metabolismo , Receptores de Interferón/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Regulación hacia Arriba/efectos de los fármacos , Receptor de Interferón gamma
16.
PLoS One ; 10(12): e0141925, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26720197

RESUMEN

The cornea is a complex tissue that must preserve its transparency to maintain optimal vision. However, in some circumstances, damage to the eye can result in neovascularization that impairs vision. This outcome can occur when herpes simplex virus type 1 (HSV-1) causes the immunoinflammatory lesion stromal keratitis (SK). Potentially useful measures to control the severity of SK are to target angiogenesis which with herpetic SK invariably involves VEGF. One such way to control angiogenesis involves the endothelial receptor Robo4 (R4), which upon interaction with another protein activates an antiangiogenic pathway that counteracts VEGF downstream signaling. In this study we show that mice unable to produce R4 because of gene knockout developed significantly higher angiogenesis after HSV-1 ocular infection than did infected wild type (WT) controls. Moreover, providing additional soluble R4 (sR4) protein by subconjunctival administration to R4 KO HSV-1 infected mice substantially rescued the WT phenotype. Finally, administration of sR4 to WT HSV-1 infected mice diminished the extent of corneal angiogenesis compared to WT control animals. Our results indicate that sR4 could represent a useful therapeutic tool to counteract corneal angiogenesis and help control the severity of SK.


Asunto(s)
Neovascularización de la Córnea/genética , Queratitis Herpética/genética , Queratitis Herpética/patología , Neovascularización Patológica/genética , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Animales , Neovascularización de la Córnea/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Herpesvirus Humano 1 , Queratitis Herpética/tratamiento farmacológico , Ratones , Ratones Noqueados , Neovascularización Patológica/tratamiento farmacológico , Proteínas del Tejido Nervioso/farmacología , Fenotipo , Receptores de Superficie Celular , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Cell ; 157(6): 1309-1323, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24906149

RESUMEN

When killer lymphocytes recognize infected cells, perforin delivers cytotoxic proteases (granzymes) into the target cell to trigger apoptosis. What happens to intracellular bacteria during this process is unclear. Human, but not rodent, cytotoxic granules also contain granulysin, an antimicrobial peptide. Here, we show that granulysin delivers granzymes into bacteria to kill diverse bacterial strains. In Escherichia coli, granzymes cleave electron transport chain complex I and oxidative stress defense proteins, generating reactive oxygen species (ROS) that rapidly kill bacteria. ROS scavengers and bacterial antioxidant protein overexpression inhibit bacterial death. Bacteria overexpressing a GzmB-uncleavable mutant of the complex I subunit nuoF or strains that lack complex I still die, but more slowly, suggesting that granzymes disrupt multiple vital bacterial pathways. Mice expressing transgenic granulysin are better able to clear Listeria monocytogenes. Thus killer cells play an unexpected role in bacterial defense.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Infecciones Bacterianas/inmunología , Escherichia coli , Leucocitos Mononucleares/inmunología , Listeria monocytogenes , Staphylococcus aureus , Animales , Granzimas/metabolismo , Células HeLa , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos BALB C , Perforina/genética , Perforina/metabolismo , Especies Reactivas de Oxígeno/metabolismo
19.
J Immunol ; 192(6): 2734-43, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24516198

RESUMEN

HSV infection of adult humans occasionally results in life-threatening herpes simplex encephalitis (HSE) for reasons that remain to be defined. An animal system that could prove useful to model HSE could be microRNA-155 knockout (miR-155KO) mice. Thus, we observe that mice with a deficiency of miR-155 are highly susceptible to HSE with a majority of animals (75-80%) experiencing development of HSE after ocular infection with HSV-1. The lesions appeared to primarily represent the destructive consequences of viral replication, and animals could be protected from HSE by acyclovir treatment provided 4 d after ocular infection. The miR-155KO animals were also more susceptible to development of zosteriform lesions, a reflection of viral replication and dissemination within the nervous system. One explanation for the heightened susceptibility to HSE and zosteriform lesions could be because miR-155KO animals develop diminished CD8 T cell responses when the numbers, functionality, and homing capacity of effector CD8 T cell responses were compared. Indeed, adoptive transfer of HSV-immune CD8 T cells to infected miR-155KO mice at 24 h postinfection provided protection from HSE. Deficiencies in CD8 T cell numbers and function also explained the observation that miR-155KO animals were less able than control animals to maintain HSV latency. To our knowledge, our observations may be the first to link miR-155 expression with increased susceptibility of the nervous system to virus infection.


Asunto(s)
Encéfalo/metabolismo , Encefalitis por Herpes Simple/genética , Predisposición Genética a la Enfermedad/genética , MicroARNs/genética , Aciclovir/farmacología , Traslado Adoptivo , Animales , Antivirales/farmacología , Encéfalo/patología , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Encefalitis por Herpes Simple/terapia , Encefalitis por Herpes Simple/virología , Femenino , Citometría de Flujo , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Supervivencia , Replicación Viral/efectos de los fármacos
20.
Invest Ophthalmol Vis Sci ; 54(9): 6269-79, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23942967

RESUMEN

PURPOSE: Neuroprotectin D1 (NPD1) is an anti-inflammatory and proresolving lipid mediator biosynthesized from the omega-3-polyunsaturated fatty acid docosahexaenoic acid (DHA). The purpose of this study is to test the therapeutic potential of NPD1 for the treatment of herpes simplex virus (HSV)-induced stromal keratitis (SK) using a mouse model. METHODS: C57BL/6 mice were infected ocularly with HSV-1 strain RE. Infected animals were treated topically with methyl ester prodrug NPD1 (300 ng/eye, 5-µL drop). Development of SK lesions, infiltration of inflammatory cells into the cornea, and production of proinflammatory cytokines, chemokines, and angiogenic factors were compared to untreated animals using slit-lamp biomicroscopy, flow cytometry, ELISA, and quantitative PCR (qPCR). RESULTS: Topical administration of NPD1 resulted in a significant reduction in the severity and incidence of SK, as well as the extent of corneal neovascularization in the NPD1-treated animals compared to their untreated counterparts. Infiltration of fewer neutrophils and pathogenic CD4⁺ T cells into the cornea, along with a lower number of cells that could be induced ex vivo to produce IFN-γ and IL-17, occurred with NPD1 treatment. Additionally, treatment with NPD1 diminished the production of proinflammatory cytokines, chemokines, and angiogenic factors, such as IL-6, CXCL1, CXCL-10, CCL-20, VEGF-A, MMP-2, and MMP-9 in the corneas of infected animals. Importantly, treatment with NPD1 increased the production of the anti-inflammatory cytokine, IL-10. CONCLUSIONS: Our novel findings demonstrate that NPD1 treatment could represent a valuable therapeutic approach to control SK lesions.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ácidos Docosahexaenoicos/uso terapéutico , Queratitis Herpética/tratamiento farmacológico , Administración Tópica , Animales , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Quimiocinas/metabolismo , Neovascularización de la Córnea/tratamiento farmacológico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inmunohistoquímica , Queratitis Herpética/inmunología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...