Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38648067

RESUMEN

We recently reported that resistance to PD-1-blockade in a refractory lung cancer-derived model involved increased collagen deposition and the collagen-binding inhibitory receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), and thus we hypothesized that LAIR1 and collagen cooperated to suppress therapeutic response. Here, we report LAIR1 is associated with tumor stroma and is highly expressed by intratumoral myeloid cells in both human tumors and mouse models of cancer. Stroma-associated myeloid cells exhibit a suppressive phenotype and correlate with LAIR1 expression in human cancer. NGM438, a novel humanized LAIR1 antagonist monoclonal antibody, elicits myeloid inflammation and allogeneic T cell responses by binding to LAIR1 and blocking collagen engagement. Further, a mouse-reactive NGM438 surrogate antibody sensitized refractory KP mouse lung tumors to anti-PD-1 therapy and resulted in increased intratumoral CD8+ T cell content and inflammatory gene expression. These data place LAIR1 at the intersection of stroma and suppressive myeloid cells and support the notion that blockade of the LAIR1/collagen axis can potentially address resistance to checkpoint inhibitor therapy in the clinic.

2.
Cancer Immunol Res ; 12(5): 592-613, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38393969

RESUMEN

Solid tumors are dense three-dimensional (3D) multicellular structures that enable efficient receptor-ligand trans interactions via close cell-cell contact. Immunoglobulin-like transcript (ILT)2 and ILT4 are related immune-suppressive receptors that play a role in the inhibition of myeloid cells within the tumor microenvironment. The relative contribution of ILT2 and ILT4 to immune inhibition in the context of solid tumor tissue has not been fully explored. We present evidence that both ILT2 and ILT4 contribute to myeloid inhibition. We found that although ILT2 inhibits myeloid cell activation in the context of trans-engagement by MHC-I, ILT4 efficiently inhibits myeloid cells in the presence of either cis- or trans-engagement. In a 3D spheroid tumor model, dual ILT2/ILT4 blockade was required for the optimal activation of myeloid cells, including the secretion of CXCL9 and CCL5, upregulation of CD86 on dendritic cells, and downregulation of CD163 on macrophages. Humanized mouse tumor models showed increased immune activation and cytolytic T-cell activity with combined ILT2 and ILT4 blockade, including evidence of the generation of immune niches, which have been shown to correlate with clinical response to immune-checkpoint blockade. In a human tumor explant histoculture system, dual ILT2/ILT4 blockade increased CXCL9 secretion, downregulated CD163 expression, and increased the expression of M1 macrophage, IFNγ, and cytolytic T-cell gene signatures. Thus, we have revealed distinct contributions of ILT2 and ILT4 to myeloid cell biology and provide proof-of-concept data supporting the combined blockade of ILT2 and ILT4 to therapeutically induce optimal myeloid cell reprogramming in the tumor microenvironment.


Asunto(s)
Antígenos CD , Receptor Leucocitario Tipo Inmunoglobulina B1 , Glicoproteínas de Membrana , Células Mieloides , Receptores Inmunológicos , Microambiente Tumoral , Receptores Inmunológicos/metabolismo , Animales , Humanos , Ratones , Microambiente Tumoral/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Glicoproteínas de Membrana/metabolismo , Línea Celular Tumoral , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo
3.
Cell Rep Med ; 4(7): 101110, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37467717

RESUMEN

Multiple myeloma (MM) is an incurable malignancy of plasma cells. To identify targets for MM immunotherapy, we develop an integrated pipeline based on mass spectrometry analysis of seven MM cell lines and RNA sequencing (RNA-seq) from 900+ patients. Starting from 4,000+ candidates, we identify the most highly expressed cell surface proteins. We annotate candidate protein expression in many healthy tissues and validate the expression of promising targets in 30+ patient samples with relapsed/refractory MM, as well as in primary healthy hematopoietic stem cells and T cells by flow cytometry. Six candidates (ILT3, SEMA4A, CCR1, LRRC8D, FCRL3, IL12RB1) and B cell maturation antigen (BCMA) present the most favorable profile in malignant and healthy cells. We develop a bispecific T cell engager targeting ILT3 that shows potent killing effects in vitro and decreased tumor burden and prolonged mice survival in vivo, suggesting therapeutic relevance. Our study uncovers MM-associated antigens that hold great promise for immune-based therapies of MM.


Asunto(s)
Mieloma Múltiple , Animales , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Inmunoterapia/métodos , Linfocitos T , Células Plasmáticas/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(41): E6172-E6181, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27681624

RESUMEN

The regulation of host-pathogen interactions during Mycobacterium tuberculosis (Mtb) infection remains unresolved. MicroRNAs (miRNAs) are important regulators of the immune system, and so we used a systems biology approach to construct an miRNA regulatory network activated in macrophages during Mtb infection. Our network comprises 77 putative miRNAs that are associated with temporal gene expression signatures in macrophages early after Mtb infection. In this study, we demonstrate a dual role for one of these regulators, miR-155. On the one hand, miR-155 maintains the survival of Mtb-infected macrophages, thereby providing a niche favoring bacterial replication; on the other hand, miR-155 promotes the survival and function of Mtb-specific T cells, enabling an effective adaptive immune response. MiR-155-induced cell survival is mediated through the SH2 domain-containing inositol 5-phosphatase 1 (SHIP1)/protein kinase B (Akt) pathway. Thus, dual regulation of the same cell survival pathway in innate and adaptive immune cells leads to vastly different outcomes with respect to bacterial containment.


Asunto(s)
Inmunidad Adaptativa/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , MicroARNs/genética , Mycobacterium tuberculosis/inmunología , Tuberculosis/genética , Tuberculosis/inmunología , Animales , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Transcriptoma , Tuberculosis/metabolismo
5.
J Exp Med ; 212(5): 715-28, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25918344

RESUMEN

Immune control of persistent infection with Mycobacterium tuberculosis (Mtb) requires a sustained pathogen-specific CD4 T cell response; however, the molecular pathways governing the generation and maintenance of Mtb protective CD4 T cells are poorly understood. Using MHCII tetramers, we show that Mtb-specific CD4 T cells are subject to ongoing antigenic stimulation. Despite this chronic stimulation, a subset of PD-1(+) cells is maintained within the lung parenchyma during tuberculosis (TB). When transferred into uninfected animals, these cells persist, mount a robust recall response, and provide superior protection to Mtb rechallenge when compared to terminally differentiated Th1 cells that reside preferentially in the lung-associated vasculature. The PD-1(+) cells share features with memory CD4 T cells in that their generation and maintenance requires intrinsic Bcl6 and intrinsic ICOS expression. Thus, the molecular pathways required to maintain Mtb-specific CD4 T cells during ongoing infection are similar to those that maintain memory CD4 T cells in scenarios of antigen deprivation. These results suggest that vaccination strategies targeting the ICOS and Bcl6 pathways in CD4 T cells may provide new avenues to prevent TB.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Memoria Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/inmunología , Inmunidad Celular/genética , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-6 , Células TH1/patología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/patología
6.
Nat Immunol ; 16(1): 67-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25419628

RESUMEN

Immune responses are tightly regulated to ensure efficient pathogen clearance while avoiding tissue damage. Here we report that Setdb2 was the only protein lysine methyltransferase induced during infection with influenza virus. Setdb2 expression depended on signaling via type I interferons, and Setdb2 repressed expression of the gene encoding the neutrophil attractant CXCL1 and other genes that are targets of the transcription factor NF-κB. This coincided with occupancy by Setdb2 at the Cxcl1 promoter, which in the absence of Setdb2 displayed diminished trimethylation of histone H3 Lys9 (H3K9me3). Mice with a hypomorphic gene-trap construct of Setdb2 exhibited increased infiltration of neutrophils during sterile lung inflammation and were less sensitive to bacterial superinfection after infection with influenza virus. This suggested that a Setdb2-mediated regulatory crosstalk between the type I interferons and NF-κB pathways represents an important mechanism for virus-induced susceptibility to bacterial superinfection.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/inmunología , FN-kappa B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Neumonía/inmunología , Sobreinfección/inmunología , Animales , Quimiocina CXCL1/inmunología , Susceptibilidad a Enfermedades , Femenino , Interferón Tipo I/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/virología , Neumonía/enzimología , Neumonía/virología , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Organismos Libres de Patógenos Específicos , Sobreinfección/enzimología , Sobreinfección/microbiología
7.
Cell Host Microbe ; 15(2): 153-63, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24528862

RESUMEN

Chronic tuberculosis in an immunocompetent host is a consequence of the delicately balanced growth of Mycobacterium tuberculosis (Mtb) in the face of host defense mechanisms. We identify an Mtb enzyme (TdmhMtb) that hydrolyzes the mycobacterial glycolipid trehalose dimycolate and plays a critical role in balancing the intracellular growth of the pathogen. TdmhMtb is induced under nutrient-limiting conditions and remodels the Mtb envelope to increase nutrient influx but concomitantly sensitizes Mtb to stresses encountered in the host. Consistent with this, a ΔtdmhMtb mutant is more resilient to stress and grows to levels higher than those of wild-type in immunocompetent mice. By contrast, mutant growth is retarded in MyD88(-/-) mice, indicating that TdmhMtb provides a growth advantage to intracellular Mtb in an immunocompromised host. Thus, the effects and countereffects of TdmhMtb play an important role in balancing intracellular growth of Mtb in a manner that is directly responsive to host innate immunity.


Asunto(s)
Factores Cordón/metabolismo , Hidrolasas/metabolismo , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/fisiología , Animales , Citosol/microbiología , Eliminación de Gen , Hidrolasas/genética , Hidrólisis , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crecimiento & desarrollo
8.
J Immunol ; 189(1): 23-7, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661094

RESUMEN

Cellular fusion of macrophages into multinucleated giant cells is a distinguishing feature of the granulomatous response to inflammation, infection, and foreign bodies (Kawai and Akira. 2011. Immunity 34: 637-650). We observed a marked increase in fusion of macrophages genetically deficient in Dicer, an enzyme required for canonical microRNA (miRNA) biogenesis. Gene expression profiling of miRNA-deficient macrophages revealed an upregulation of the IL-4-responsive fusion protein Tm7sf4, and analyses identified miR-7a-1 as a negative regulator of macrophage fusion, functioning by directly targeting Tm7sf4 mRNA. miR-7a-1 is itself an IL-4-responsive gene in macrophages, suggesting feedback control of cellular fusion. Collectively, these data indicate that miR-7a-1 functions to regulate IL-4-directed multinucleated giant cell formation.


Asunto(s)
Diferenciación Celular/inmunología , Células Gigantes de Langhans/inmunología , Macrófagos/citología , Macrófagos/inmunología , MicroARNs/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Fusión Celular/métodos , Células Cultivadas , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Células Gigantes de Langhans/citología , Células Gigantes de Langhans/metabolismo , Células HEK293 , Humanos , Interleucina-4/fisiología , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , Ribonucleasa III/deficiencia , Ribonucleasa III/genética , Transcripción Genética/inmunología
9.
J Clin Microbiol ; 44(7): 2595-600, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16825391

RESUMEN

Normal human serum inhibits Acanthamoeba (encephalitis isolate) binding to and cytotoxicity of human brain microvascular endothelial cells, which constitute the blood-brain barrier. Zymographic assays revealed that serum inhibits extracellular protease activities of acanthamoebae. But it is most likely that inhibition of specific properties of acanthamoebae is a consequence of the initial amoebicidal-amoebistatic effects induced by serum. For example, serum exhibited amoebicidal effects; i.e., up to 50% of the exposed trophozoites were killed. The residual subpopulation, although viable, remained static over longer incubations. Interestingly, serum enhanced the phagocytic ability of acanthamoebae, as measured by bacterial uptake. Overall, our results demonstrate that human serum has inhibitory effects on Acanthamoeba growth and viability, protease secretions, and binding to and subsequent cytotoxicity for brain microvascular endothelial cells. Conversely, Acanthamoeba phagocytosis was stimulated by serum.


Asunto(s)
Acanthamoeba castellanii/fisiología , Suero/fisiología , Acanthamoeba castellanii/crecimiento & desarrollo , Acanthamoeba castellanii/metabolismo , Acanthamoeba castellanii/patogenicidad , Encéfalo/irrigación sanguínea , Adhesión Celular , Muerte Celular , Células Endoteliales/citología , Células Endoteliales/parasitología , Humanos , Péptido Hidrolasas/metabolismo , Fagocitosis , Proteínas Protozoarias/biosíntesis
10.
BMC Microbiol ; 6: 42, 2006 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-16672059

RESUMEN

BACKGROUND: Granulomatous amoebic encephalitis due to Acanthamoeba is often a fatal human disease. However, the pathogenesis and pathophysiology of Acanthamoeba encephalitis remain unclear. In this study, the role of extracellular Acanthamoeba proteases in central nervous system pathogenesis and pathophysiology was examined. RESULTS: Using an encephalitis isolate belonging to T1 genotype, we observed two major proteases with approximate molecular weights of 150 KD and 130 KD on SDS-PAGE gels using gelatin as substrate. The 130 KD protease was inhibited with phenylmethylsulfonyl fluoride (PMSF) suggesting that it is a serine protease, while the 150 KD protease was inhibited with 1, 10-phenanthroline suggesting that it is a metalloprotease. Both proteases exhibited maximal activity at neutral pH and over a range of temperatures, indicating their physiological relevance. These proteases degrade extracellular matrix (ECM), which provide structural and functional support to the brain tissue, as shown by the degradation of collagen I and III (major components of collagenous ECM), elastin (elastic fibrils of ECM), plasminogen (involved in proteolytic degradation of ECM), as well as casein and haemoglobin. The proteases were purified partially using ion-exchange chromatography and their effects were tested in an in vitro model of the blood-brain barrier using human brain microvascular endothelial cells (HBMEC). Neither the serine nor the metalloprotease exhibited HBMEC cytotoxicity. However, the serine protease exhibited HBMEC monolayer disruptions (trypsin-like) suggesting a role in blood-brain barrier perturbations. CONCLUSION: Overall, these data suggest that Acanthamoeba proteases digest ECM, which may play crucial role(s) in invasion of the brain tissue by amoebae.


Asunto(s)
Acanthamoeba/enzimología , Acanthamoeba/patogenicidad , Amebiasis/parasitología , Infecciones Protozoarias del Sistema Nervioso Central/parasitología , Encefalitis/parasitología , Metaloproteasas/metabolismo , Serina Endopeptidasas/metabolismo , Acanthamoeba/clasificación , Acanthamoeba/genética , Queratitis por Acanthamoeba/parasitología , Queratitis por Acanthamoeba/fisiopatología , Amebiasis/fisiopatología , Animales , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Células Cultivadas , Infecciones Protozoarias del Sistema Nervioso Central/fisiopatología , Encefalitis/fisiopatología , Endotelio Vascular/citología , Matriz Extracelular/metabolismo , Humanos , Metaloproteasas/química , Microcirculación , Serina Endopeptidasas/química
11.
J Med Microbiol ; 55(Pt 6): 689-694, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16687585

RESUMEN

The ability of Acanthamoeba to feed on Gram-negative bacteria, as well as to harbour potential pathogens, such as Legionella pneumophila, Coxiella burnetii, Pseudomonas aeruginosa, Vibrio cholerae, Helicobacter pylori, Listeria monocytogenes and Mycobacterium avium, suggest that both amoebae and bacteria are involved in complex interactions, which may play important roles in the environment and in human health. In this study, Acanthamoeba castellanii (a keratitis isolate belonging to the T4 genotype) was used and its interactions with Escherichia coli (strain K1, a cerebrospinal fluid isolate from a meningitis patient, O18 : K1 : H7, and a K-12 laboratory strain, HB101) were studied. The invasive K1 isolate exhibited a significantly higher association with A. castellanii than the non-invasive K-12 isolate. Similarly, K1 showed significantly increased invasion and/or uptake by A. castellanii in gentamicin protection assays than the non-invasive K-12. Using several mutants derived from K1, it was observed that outer-membrane protein A (OmpA) and LPS were crucial bacterial determinants responsible for E. coli K1 interactions with A. castellanii. Once inside the cell, E. coli K1 remained viable and multiplied within A. castellanii, while E. coli K-12 was killed. Again, OmpA and LPS were crucial for E. coli K1 intracellular survival in A. castellanii. In conclusion, these findings suggest that E. coli K1 interactions with A. castellanii are carefully regulated by the virulence of E. coli.


Asunto(s)
Acanthamoeba castellanii/fisiología , Escherichia coli/fisiología , Acanthamoeba castellanii/genética , Acanthamoeba castellanii/aislamiento & purificación , Acanthamoeba castellanii/patogenicidad , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/fisiología , Microbiología Ambiental , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Escherichia coli/patogenicidad , Escherichia coli K12/genética , Escherichia coli K12/patogenicidad , Escherichia coli K12/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiología , Humanos , Técnicas In Vitro , Lipopolisacáridos/metabolismo , Especificidad de la Especie , Simbiosis/genética , Simbiosis/fisiología , Virulencia/genética , Virulencia/fisiología
12.
J Med Microbiol ; 54(Pt 8): 755-759, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16014429

RESUMEN

The majority of the keratitis-causing Acanthamoeba isolates are genotype T4. In an attempt to determine whether predominance of T4 isolates in Acanthamoeba keratitis is due to greater virulence or greater prevalence of this genotype, Acanthamoeba genotypes were determined for 13 keratitis isolates and 12 environmental isolates from Iran. Among 13 clinical isolates, eight (61.5%) belonged to T4, two (15.3%) belonged to T3 and three (23%) belonged to the T2 genotype. In contrast, the majority of 12 environmental isolates tested in the present study belonged to T2 (7/12, 58.3%), followed by 4/12 T4 isolates (33.3%). In addition, the genotypes of six new Acanthamoeba isolates from UK keratitis cases were determined. Of these, five (83.3%) belonged to T4 and one was T3 (16.6%), supporting the expected high frequency of T4 in Acanthamoeba keratitis. In total, the genotypes of 24 Acanthamoeba keratitis isolates from the UK and Iran were determined. Of these, 17 belonged to T4 (70.8%), three belonged to T2 (12.5%), three belonged to T3 (12.5%) and one belonged to T11 (4.1%), confirming that T4 is the predominant genotype (S2=4.167; P=0.0412) in Acanthamoeba keratitis.


Asunto(s)
Acanthamoeba/aislamiento & purificación , Acanthamoeba/fisiología , Acanthamoeba/genética , Acanthamoeba/patogenicidad , Queratitis por Acanthamoeba/parasitología , Animales , Genotipo , Irán , ARN Ribosómico 18S/análisis , ARN Ribosómico 18S/genética , Análisis de Secuencia de ADN , Reino Unido
13.
Microbes Infect ; 7(13): 1345-51, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16027019

RESUMEN

Balamuthia mandrillaris is an emerging protozoan parasite that can cause fatal granulomatous encephalitis. Haematogenous spread is a likely route prior to entry into the central nervous system (CNS), but it is not clear how circulating amoebae cross the blood-brain barrier. Using human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier, we determined HBMEC inflammatory response to B. mandrillaris and the underlying mechanisms associated with this response. We demonstrated that HBMEC incubated with B. mandrillaris released significantly higher levels of interleukin-6 (IL-6) (>400 pg/ml) as compared with less than 50 pg/ml in HBMEC incubated alone. Western blotting assays determined that B. mandrillaris specifically activates phosphatidylinositol 3-kinase (PI3K). By using LY294002, a PI3K inhibitor, as well as by using HBMEC expressing dominant-negative PI3K, we have identified PI3K as an important mediator of B. mandrillaris-mediated IL-6 release. We conclude that B. mandrillaris induces HBMEC signalling pathways, which lead to IL-6 release. This is the first time PI3K has been shown to play a crucial role in B. mandrillaris-mediated IL-6 release in HBMEC.


Asunto(s)
Infecciones Protozoarias del Sistema Nervioso Central/parasitología , Células Endoteliales/parasitología , Interleucina-6/inmunología , Lobosea/patogenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Barrera Hematoencefálica , Células Cultivadas , Infecciones Protozoarias del Sistema Nervioso Central/inmunología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Humanos , Lobosea/inmunología , Fosfatidilinositol 3-Quinasas/fisiología
14.
J Infect ; 51(2): 150-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16038767

RESUMEN

OBJECTIVES: Granulomatous amoebic encephalitis (GAE) is a serious human infection with fatal consequences, however, the pathogenic mechanisms associated with this disease remain unclear. Several lines of evidence suggest that haematogenous spread is a prerequisite for Acanthamoeba encephalitis but it is not clear how circulating amoebae cross the blood-brain barrier to gain entry into the central nervous system. Objectives of this study were to determine the effects of Acanthamoeba on the permeability of an in vitro blood-brain barrier model and factors contributing to these changes. METHODS: Using human brain microvascular endothelial cells, an in vitro blood-brain barrier model was constructed in 24-well Transwell plates. Acanthamoeba (GAE isolate belonging to T1 genotype) or its conditioned media were used to determine permeability changes. Zymography assays were performed to characterise Acanthamoeba proteases. In addition, the ability of Acanthamoeba to bind brain microvascular endothelial cells was determined using adhesion assays. RESULTS: We observed that Acanthamoeba produced an increase of more than 45% in the blood-brain barrier permeability. Acanthamoeba-conditioned media exhibited similar effects indicating Acanthamoeba-mediated blood-brain barrier permeability is contact-independent. Prior treatment of conditioned media with phenylmethyl sulfonyl fluoride (PMSF, serine protease inhibitor), abolished permeability changes indicating the role of serine proteases. Of interest, methyl alpha-d-mannopyranoside inhibited Acanthamoeba binding to human brain microvascular endothelial cells but had no effect on Acanthamoeba-mediated blood-brain barrier permeability. Zymography assays revealed that Acanthamoeba produced two major proteases, one of which was inhibited by PMSF (serine protease inhibitor) and the second with 1,10-phenanthroline (metalloprotease inhibitor). CONCLUSIONS: We have for the first time shown that Acanthamoeba produces human brain microvascular endothelial cells permeability, which can be blocked by PMSF. A metalloprotease of approx. molecular weight of 150 kDa is produced by A. castellanii (GAE isolate belonging to T1 genotype) and its role in the disease is suggested.


Asunto(s)
Acanthamoeba castellanii/enzimología , Acanthamoeba castellanii/patogenicidad , Barrera Hematoencefálica/metabolismo , Metaloproteasas/fisiología , Serina Endopeptidasas/fisiología , Acanthamoeba castellanii/genética , Amebiasis/parasitología , Amebiasis/fisiopatología , Animales , Barrera Hematoencefálica/parasitología , Adhesión Celular/fisiología , Línea Celular , Medios de Cultivo Condicionados , Encefalitis/parasitología , Encefalitis/fisiopatología , Células Endoteliales/parasitología , Células Endoteliales/fisiología , Genotipo , Humanos , Lectina de Unión a Manosa/fisiología , Metaloproteasas/metabolismo , Péptido Hidrolasas/metabolismo , Permeabilidad , Serina Endopeptidasas/metabolismo , Factores de Virulencia/clasificación
15.
Acta Trop ; 95(2): 100-8, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15963936

RESUMEN

Acanthamoeba is an opportunistic protozoan that is widely distributed in the environment and can cause human infections. The life cycle of Acanthamoeba consists of an infective trophozoite form. However under harsh environmental conditions trophozoites differentiate into a double-walled, metabolically inactive and resistant cyst form. Research in Acanthamoeba has mostly focussed on the infective trophozoite form and its pathogenic mechanisms. In this study, we used Acanthamoeba isolates belonging to T1, T2, T3, T4, T7 genotypes and studied their cysts properties. We determined that food deprivation stimulates encystment in Acanthamoeba isolates belonging to T1, T2, T3, T4 and T7 genotypes in a sodium dodecyl sulfate (SDS)-resistant manner. In addition, increase in osmolarity triggered encystment in T1, T2, T3, T4 isolates (SDS-resistant) but T7 failed to encyst (SDS-labile). Adhesion assays revealed that Acanthamoeba cysts belonging to T1, T2, T3, T4, and T7 genotypes exhibited no and/or minimal binding (<5%) to the host cells. Fluorescein-labelled lectins showed that all Acanthamoeba isolates tested exhibited binding to concanavalin A, indicating the expression of mannosyl- and/or glucosyl-residues. Role of cysts in the transmission of infection is discussed further.


Asunto(s)
Acanthamoeba/clasificación , Epitelio Corneal/citología , Acanthamoeba/genética , Acanthamoeba/crecimiento & desarrollo , Animales , Adhesión Celular , Células Cultivadas , Epitelio Corneal/parasitología , Genotipo , Humanos , Estadios del Ciclo de Vida , Concentración Osmolar
16.
Parasitol Res ; 96(6): 402-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15940518

RESUMEN

Using fluorescein isothiocyanate (FITC)-labelled Escherichia coli, phagocytosis in Acanthamoeba is studied. This assay is based on the quenching effect of trypan blue on FITC-labelled E. coli. Only intracellular E. coli retain their fluorescence, which are easily discriminated from non-fluorescent adherent bacteria. Acanthamoeba uptake of E. coli is significantly reduced in the presence of genistein, a protein tyrosine kinase inhibitor. In contrast, sodium orthovanadate (protein tyrosine phosphatase inhibitor) increases bacterial uptake by Acanthamoeba. Treatment of Acanthamoeba with cytochalasin D (actin polymerization inhibitor) abolished the ability of Acanthamoeba to phagocytose E. coli suggesting that tyrosine kinase-mediated signaling may play a role in Acanthamoeba phagocytosis. In addition, we showed that phosphatidylinositol 3-kinase (PI3K) plays an important role in Acanthamoeba uptake of E. coli. Role of mannose-binding protein in Acanthamoeba phagocytosis is discussed further.


Asunto(s)
Acanthamoeba castellanii/enzimología , Enzimas/metabolismo , Fagocitosis/fisiología , Acanthamoeba castellanii/efectos de los fármacos , Acanthamoeba castellanii/microbiología , Animales , Citocalasina D/farmacología , Inhibidores Enzimáticos/farmacología , Escherichia coli/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Genisteína/farmacología , Fagocitosis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Azul de Tripano/metabolismo , Vanadatos/farmacología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
17.
Infect Immun ; 73(5): 2704-8, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15845472

RESUMEN

Granulomatous amoebic encephalitis due to Acanthamoeba castellanii is a serious human infection with fatal consequences, but it is not clear how the circulating amoebae interact with the blood-brain barrier and transmigrate into the central nervous system. We studied the effects of an Acanthamoeba encephalitis isolate belonging to the T1 genotype on human brain microvascular endothelial cells, which constitute the blood-brain barrier. Using an apoptosis-specific enzyme-linked immunosorbent assay, we showed that Acanthamoeba induces programmed cell death in brain microvascular endothelial cells. Next, we observed that Acanthamoeba specifically activates phosphatidylinositol 3-kinase. Acanthamoeba-mediated brain endothelial cell death was abolished using LY294002, a phosphatidylinositol 3-kinase inhibitor. These results were further confirmed using brain microvascular endothelial cells expressing dominant negative forms of phosphatidylinositol 3-kinase. This is the first demonstration that Acanthamoeba-mediated brain microvascular endothelial cell death is dependent on phosphatidylinositol 3-kinase.


Asunto(s)
Acanthamoeba castellanii/patogenicidad , Apoptosis , Encéfalo/irrigación sanguínea , Células Endoteliales/parasitología , Microcirculación/parasitología , Fosfatidilinositol 3-Quinasas/metabolismo , Amebiasis/parasitología , Amebiasis/fisiopatología , Animales , Barrera Hematoencefálica , Células Cultivadas , Infecciones Protozoarias del Sistema Nervioso Central/parasitología , Infecciones Protozoarias del Sistema Nervioso Central/fisiopatología , Encefalitis/parasitología , Encefalitis/fisiopatología , Células Endoteliales/enzimología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/parasitología , Humanos , Microcirculación/enzimología , Microcirculación/fisiología
18.
Microb Pathog ; 37(5): 231-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15519044

RESUMEN

Acanthamoeba are opportunistic protozoan parasites that can cause fatal granulomatous amoebic encephalitis and eye keratitis, however the pathogenic mechanisms of Acanthamoeba remain unclear. In this study, we described the ability of live Acanthamoeba to hydrolyse extracellular ATP. Both clinical and non-clinical isolates belonging to genotypes, T1, T2, T3, T4 and T7 exhibited ecto-ATPase activities in vitro. Using non-denaturing polyacrylamide gel electrophoresis, ecto-ATPases were further characterized. All Acanthamoeba isolates tested, exhibited a single ecto-ATPase band (approximate molecular weight of 272 kDa). However, clinical isolates exhibited additional bands suggesting that ecto-ATPases may play a role in the pathogenesis of Acanthamoeba. This was supported using suramin (ecto-ATPase inhibitor), which inhibited Acanthamoeba-induced host cell cytotoxicity. Previously, we and others have shown that Acanthamoeba binds to host cells using their mannose-binding protein and binding can be blocked using exogenous alpha-mannose. In this study, we observed that alpha-mannose significantly increased ecto-ATPase activities of pathogenic Acanthamoeba belonging to T1, T2, T3 and T4 genotypes but had no effect on non-pathogenic Acanthamoeba (belonging to T7 genotype). Overall, we have shown, for the first time, that Acanthamoeba exhibit ecto-ATPase activities, which may play a role in the pathogenesis of Acanthamoeba as well as their potential role in the differentiation of pathogenic Acanthamoeba.


Asunto(s)
Acanthamoeba/enzimología , Adenosina Trifosfatasas/metabolismo , Acanthamoeba/genética , Adenosina Trifosfatasas/genética , Animales , Adhesión Celular , Células Epiteliales/citología , Células Epiteliales/parasitología , Epitelio Corneal/citología , Epitelio Corneal/parasitología , Regulación de la Expresión Génica , Humanos , Hidrólisis , Manosa/metabolismo , Lectina de Unión a Manosa/metabolismo , Suramina/farmacología
19.
J Med Microbiol ; 53(Pt 10): 1007-1012, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15358823

RESUMEN

The first isolation in the UK of Balamuthia mandrillaris amoebae from a fatal case of granulomatous amoebic meningoencephalitis is reported. Using primary cultures of human brain microvascular endothelial cells (HBMECs), amoebae were isolated from the brain and cerebrospinal fluid (CSF). The cultures showed a cytopathic effect at 20-28 days, but morphologically identifiable B. mandrillaris amoebae were seen in cleared plaques in subcultures at 45 days. The identification of the organism was later confirmed using PCR on Chelex-treated extracts. Serum taken while the patient was still alive reacted strongly with slide antigen prepared from cultures of the post-mortem isolate, and also with those from a baboon B. mandrillaris strain at 1:10,000 in indirect immunofluorescence, but with Acanthamoeba castellanii (Neff) at 1:160, supporting B. mandrillaris to be the causative agent. If the presence of amoebae in the post-mortem CSF reflects the condition in life, PCR studies on CSF and on biopsies of cutaneous lesions may also be a valuable tool. The role of HBMECs in understanding the interactions of B. mandrillaris with the blood-brain barrier is discussed.


Asunto(s)
Amebiasis/parasitología , Amoeba/aislamiento & purificación , Encéfalo/parasitología , Infecciones Protozoarias del Sistema Nervioso Central/parasitología , Líquido Cefalorraquídeo/parasitología , Células Endoteliales/parasitología , Granuloma/parasitología , Animales , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Humanos , Papio , Reacción en Cadena de la Polimerasa
20.
J Med Microbiol ; 53(Pt 8): 711-717, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15272056

RESUMEN

Acanthamoeba can cause fatal granulomatous amoebic encephalitis (GAE) and eye keratitis. However, the pathogenesis and pathophysiology of these emerging diseases remain unclear. In this study, the effects of Acanthamoeba on the host cell cycle using human brain microvascular endothelial cells (HBMEC) and human corneal epithelial cells (HCEC) were determined. Two isolates of Acanthamoeba belonging to the T1 genotype (GAE isolate) and T4 genotype (keratitis isolate) were used, which showed severe cytotoxicity on HBMEC and HCEC, respectively. No tissue specificity was observed in their ability to exhibit binding to the host cells. To determine the effects of Acanthamoeba on the host cell cycle, a cell-cycle-specific gene array was used. This screened for 96 genes specific for host cell-cycle regulation. It was observed that Acanthamoeba inhibited expression of genes encoding cyclins F and G1 and cyclin-dependent kinase 6, which are proteins important for cell-cycle progression. Moreover, upregulation was observed of the expression of genes such as GADD45A and p130 Rb, associated with cell-cycle arrest, indicating cell-cycle inhibition. Next, the effect of Acanthamoeba on retinoblastoma protein (pRb) phosphorylation was determined. pRb is a potent inhibitor of G1-to-S cell-cycle progression; however, its function is inhibited upon phosphorylation, allowing progression into S phase. Western blotting revealed that Acanthamoeba abolished pRb phosphorylation leading to cell-cycle arrest at the G1-to-S transition. Taken together, these studies demonstrated for the first time that Acanthamoeba inhibits the host cell cycle at the transcriptional level, as well as by modulating pRb phosphorylation using host cell-signalling mechanisms. A complete understanding of Acanthamoeba-host cell interactions may help in developing novel strategies to treat Acanthamoeba infections.


Asunto(s)
Acanthamoeba/patogenicidad , Ciclo Celular , Células Endoteliales/parasitología , Epitelio Corneal/parasitología , Acanthamoeba/aislamiento & purificación , Animales , Adhesión Celular/fisiología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Células Cultivadas , Ciclina G , Ciclina G1 , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/biosíntesis , Quinasas Ciclina-Dependientes/genética , Ciclinas/biosíntesis , Ciclinas/genética , Células Endoteliales/citología , Células Epiteliales/citología , Células Epiteliales/parasitología , Epitelio Corneal/citología , Fase G1 , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Fosforilación , Biosíntesis de Proteínas , Proteínas/genética , Proteína de Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Fase S , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA