Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Gastroenterology ; 160(4): 1118-1130.e3, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33307026

RESUMEN

BACKGROUND & AIMS: The effect of pregnancy on inflammatory bowel disease (IBD) remains poorly understood. We aimed to monitor intestinal inflammation using fecal calprotectin (FC) in pregnant women and their babies during early life. METHODS: Pregnant women with or without IBD and their infants were prospectively enrolled. FC levels were measured at each trimester of pregnancy and in babies throughout the first 3 years of life. Repeated-measures analysis was applied to investigate changes in FC levels while adjusting for confounders. The FC levels were correlated with the bacterial abundance in both mothers and babies. RESULTS: Six hundred and fourteen fecal samples from 358 mothers (98 with IBD) and 1005 fecal samples from 289 infants (76 born to IBD mothers) were analyzed. Pregnant Patients with IBD maintained higher FC levels through pregnancy compared with controls (P = 7.5 × 10-54). FC gradually increased in controls and declined in Patients with IBD throughout pregnancy (P for interaction = 5.8 × 10-7). Babies born to mothers with IBD presented with significantly higher FC levels than those born to controls up to 3 years of age, after adjusting for sex, delivery mode, feeding behavior, and antibiotics exposure (2 weeks to 3 months of age, P = .015; 12-36 months of age, P = .00003). Subdoligranulum, Roseburia, Fusicatenibacter, and Alistipes negatively correlated, and Streptococcus, Prevotella, Escherichia-Shigella, and Bifidobacterium positively correlated with maternal FC levels at T3. Faecalibacterium, Bifidobacterium, and Alistipes showed negative correlations, and Streptococcus were positively correlated with FC levels within 3 months of birth. CONCLUSIONS: Pregnancy is associated with decreased inflammatory activity in mothers with IBD. Higher FC levels in babies born to mothers with IBD suggest subclinical inflammation in early life, the long-term consequences of which are uncertain.


Asunto(s)
Colitis Ulcerosa/diagnóstico , Enfermedad de Crohn/diagnóstico , Complejo de Antígeno L1 de Leucocito/análisis , Complicaciones del Embarazo/diagnóstico , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Adulto , Antibacterianos/administración & dosificación , Bacterias/efectos de los fármacos , Bacterias/inmunología , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , Preescolar , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Colonoscopía , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Heces/química , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Humanos , Lactante , Recién Nacido , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Estudios Longitudinales , Masculino , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Estudios Prospectivos , Índice de Severidad de la Enfermedad
2.
Dig Dis Sci ; 67(11): 5149-5157, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35119598

RESUMEN

BACKGROUND: Mother-to-child transmission of Helicobacter pylori (H. pylori) is the primary source of intrafamilial spread in early childhood in regions of high H. pylori prevalence. However, early-in-life H. pylori colonization and associated protective or risk factors have not been fully evaluated in lower prevalence regions, such as the USA. AIMS: Therefore, from a well-characterized prospective US cohort, we selected women who provided fecal samples during pregnancy and had paired fecal samples from their babies up to 24 months postpartum. We evaluated maternal and baby factors associated with likelihood of H. pylori colonization in the babies. Fecal antigen testing was used to determine H. pylori status. We also evaluated the association between maternal breastmilk cytokines and H. pylori colonization in breastfed babies. RESULTS: Among included mother-baby pairs (n = 66), H. pylori prevalence was 31.8% in mothers and 19.7% in their babies. Dominant breastfeeding (adjusted odds ratio [aOR] 0.17, 95% CI 0.03-0.98) and maternal IBD (aOR 0.05, 95% CI 0.01-0.27) were associated with significantly lower likelihood of H. pylori colonization among babies; no other clinical factors were associated with H. pylori colonization in the babies. Matrix metalloproteinase-10 (MMP-10) and tumor necrosis factor-related activation-induced cytokine expression were significantly higher in breastmilk of mothers with H. pylori positive vs negative babies. CONCLUSIONS: Consistent with data from high H. pylori prevalence regions, our findings suggest dominant breastfeeding may protect against early H. pylori colonization. Downregulation of pro-inflammatory cytokines such as MMP-10 may be relevant in mediating this protection among breastfed babies, but more data are needed.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Femenino , Humanos , Lactante , Embarazo , Lactancia Materna , Infecciones por Helicobacter/epidemiología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Metaloproteinasa 10 de la Matriz , Estudios Prospectivos , Ligando RANK
3.
Gut ; 69(1): 42-51, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31036757

RESUMEN

BACKGROUND AND AIMS: Prenatal and early life bacterial colonisation is thought to play a major role in shaping the immune system. Furthermore, accumulating evidence links early life exposures to the risk of developing IBD later in life. We aimed to assess the effect of maternal IBD on the composition of the microbiome during pregnancy and on the offspring's microbiome. METHODS: We prospectively examined the diversity and taxonomy of the microbiome of pregnant women with and without IBD and their babies at multiple time points. We evaluated the role of maternal IBD diagnosis, the mode of delivery, antibiotic use and feeding behaviour on the microbiome composition during early life. To assess the effects of IBD-associated maternal and infant microbiota on the enteric immune system, we inoculated germ-free mice (GFM) with the respective stool and profiled adaptive and innate immune cell populations in the murine intestines. RESULTS: Pregnant women with IBD and their offspring presented with lower bacterial diversity and altered bacterial composition compared with control women and their babies. Maternal IBD was the main predictor of the microbiota diversity in the infant gut at 7, 14, 30, 60 and 90 days of life. Babies born to mothers with IBD demonstrated enrichment in Gammaproteobacteria and depletion in Bifidobacteria. Finally, GFM inoculated with third trimester IBD mother and 90-day infant stools showed significantly reduced microbial diversity and fewer class-switched memory B cells and regulatory T cells in the colon. CONCLUSION: Aberrant gut microbiota composition persists during pregnancy with IBD and alters the bacterial diversity and abundance in the infant stool. The dysbiotic microbiota triggered abnormal imprinting of the intestinal immune system in GFM.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Complicaciones del Embarazo/microbiología , Efectos Tardíos de la Exposición Prenatal/microbiología , Inmunidad Adaptativa , Adulto , Animales , Bacterias/clasificación , Bacterias/aislamiento & purificación , Disbiosis/inmunología , Disbiosis/microbiología , Trasplante de Microbiota Fecal/métodos , Heces/microbiología , Femenino , Estudios de Seguimiento , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Humanos , Recién Nacido , Enfermedades Inflamatorias del Intestino/inmunología , Masculino , Intercambio Materno-Fetal , Embarazo , Complicaciones del Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Estudios Prospectivos
4.
Glia ; 62(6): 999-1013, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24659539

RESUMEN

Treatment of cultures with toll-like receptor (TLR) ligands or cytokines has become a popular approach to investigate astrocyte neuroinflammatory responses and to simulate the neural environment in various CNS disorders. However, despite much effort, the mechanism of astrocyte activation such as their responses to the TLR ligands and IL-1 remain highly debated. We compared highly pure primary mouse and human astrocyte cultures in their ability to produce proinflammatory mediators (termed "A1") and immunoregulatory mediators (termed "A2") in response to LPS, poly IC, and IL-1 stimulation. In human astrocytes, IL-1 induced both A1 and A2 responses, poly IC induced mostly A2, and LPS induced neither. In mouse astrocytes, LPS induced mostly an A1-predominant response, poly IC induced both A1 and A2, and IL-1 neither. In addition, mouse astrocytes produce abundant IL-1 protein, whereas human astrocytes did not, despite robust IL-1 mRNA expression. Of the TLR4 receptor complex proteins, human astrocytes expressed TLR4 and MD2 but not CD14, whereas mouse astrocytes expressed all three. Mouse astrocyte CD14 (cell-associated and soluble) was potently upregulated by LPS. Silencing TLR4 or CD14 by siRNA suppressed LPS responses in mouse astrocytes. In vivo, astrocytes in LPS-injected mouse brains also expressed CD14. Our results show striking differences between human and mouse astrocytes in the use of TLR/IL-1R and subsequent downstream signaling and immune activation. IL-1 translational block in human astrocytes may be a built-in mechanism to prevent autocrine and paracrine cell activation and neuroinflammation. These results have important implications for translational research of human CNS diseases.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Interleucina-1/toxicidad , Receptores de Lipopolisacáridos/fisiología , Lipopolisacáridos/toxicidad , Animales , Animales Recién Nacidos , Células Cultivadas , Feto , Humanos , Ratones , Ratones Endogámicos C57BL , Especificidad de la Especie
5.
J Neurooncol ; 113(2): 185-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23512614

RESUMEN

Glioblastoma multiforme (GBM) is the most common, highly malignant primary tumor of the brain with poor prognosis. Even with the improved therapy regimen including temozolomide, the average survival rate is less than 2 years. Additional approaches to therapy targeting multiple aspects of glioma progression are in need. In the present work, we have tested the possibility that upregulation of the transcription factor interferon regulatory factor 3 (IRF3) can inhibit glioma invasiveness, proliferation and production of pro-inflammatory and pro-angiogenic factors in cultures of malignant glioma cell lines (U271, U87 and SNB-19). IRF3 is an essential transcription factor involved in TLR3/4-mediated signaling and generation of type I interferons. Although IRF3 has been suggested as a potential tumor suppressor gene, its role in glioma remains uninvestigated. In this study, we find that human glioma immune activation is potently elicited by a cytokine combination, IL-1/IFNγ (or poly IC), but not by bacterial lipopolysaccharide (LPS), similar to primary human astrocytes. GBM biopsy specimens show little detectable IRF3 immunoreactivity, and in vitro adenovirus-mediated IRF3 gene transfer in glioma cells modulates IL-1/IFNγ-induced cytokine and chemokine genes, resulting in upregulation of IFNß and IP-10 (IRF3-stimulated genes) and downregulation of proinflammatory and angiogenic genes including IL-8, TNFα and VEGF (IRF3-represssed genes). Cytokines (IL-1ß and TNFα) also induce the expression of miR-155 and miR-155*, the microRNAs crucial in immunity and inflammation-induced oncogenesis and this is dose-dependently suppressed by IRF3. Importantly, IRF3 also inhibits glioma proliferation, migration and invasion. Together, these data suggest that IRF3 can suppress glioma progression. Agents that promote IRF3 activation and expression (such as IRF3 gene transfer) could be explored as potential future therapy.


Asunto(s)
Neoplasias Encefálicas/patología , Movimiento Celular , Proliferación Celular , Glioma/patología , Mediadores de Inflamación/metabolismo , Inflamación/patología , Factor 3 Regulador del Interferón/metabolismo , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Adhesión Celular , Citocinas/genética , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/genética , MicroARNs/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Cicatrización de Heridas
6.
Am J Pathol ; 179(3): 1360-72, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21855684

RESUMEN

Tryptophan metabolism by the kynurenine pathway (KP) is important to the pathogenesis of inflammatory, infectious, and degenerative diseases. The 3-hydroxykynurenine (3-HK) branch of the KP is activated in macrophages and microglia, leading to the generation of 3-HK, 3-hydroxyanthranilic acid (3-HAA), and quinolinic acid, which are considered neurotoxic owing to their free radical-generating and N-methyl-d-aspartic acid receptor agonist activities. We investigated the role of 3-HAA in inflammatory and antioxidant gene expression and neurotoxicity in primary human fetal central nervous system cultures treated with cytokines (IL-1 with or without interferon-γ) or with Toll-like receptor ligands mimicking the proinflammatory central nervous system environment. Results were analyzed by microarray, Western blot, immunostain, enzyme-linked immunosorbent assay, and neurotoxicity assays. 3-HAA suppressed glial cytokine and chemokine expression and reduced cytokine-induced neuronal death. 3-HK also suppressed cytokine-induced neuronal death. Unexpectedly, 3-HAA was highly effective in inducing in astrocytes the expression of hemeoxygenase-1 (HO-1), an antioxidant enzyme with anti-inflammatory and cytoprotective properties. Optimal induction of HO-1 required 3-HAA and cytokines. In human microglia, 3-HAA weakly induced HO-1 and lipopolysaccharide suppressed microglial HO-1 expression. 3-HAA-mediated HO-1 expression was confirmed in cultured adult human astrocytes and in vivo after 3-HAA injection to mouse brains. Together, our results demonstrate the novel neuroprotective activity of the tryptophan metabolite 3-HAA and have implications for future therapeutic approaches for neuroinflammatory disorders.


Asunto(s)
Ácido 3-Hidroxiantranílico/farmacología , Antiinflamatorios/farmacología , Hemo-Oxigenasa 1/metabolismo , Nootrópicos/farmacología , Ácido 3-Hidroxiantranílico/metabolismo , Adulto , Animales , Astrocitos/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Interferón gamma/farmacología , Interleucina-1/farmacología , Quinurenina/análogos & derivados , Quinurenina/metabolismo , Ratones , Microglía/metabolismo , Neuronas/efectos de los fármacos
7.
Glia ; 59(12): 1911-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22170100

RESUMEN

Astrocytes, together with microglia and macrophages, participate in innate inflammatory responses in the CNS. Although inflammatory mediators such as interferons generated by astrocytes may be critical in the defense of the CNS, sustained unopposed cytokine signaling could result in harmful consequences. Interferon regulatory factor 3 (IRF3) is a transcription factor required for IFNß production and antiviral immunity. Most cells express low levels of IRF3 protein, and the transcriptional mechanism that upregulates IRF3 expression is not known. In this study, we explored the consequence of adenovirus-mediated IRF3 gene transfer (Ad-IRF3) in primary human astrocytes. We show that IRF3 transgene expression suppresses proinflammatory cytokine gene expression upon challenge with IL-1/IFNγ and alters astrocyte activation phenotype from a proinflammatory to an anti-inflammatory one, akin to an M1-M2 switch in macrophages. This was accompanied by the rescue of neurons from cytokine-induced death in glial-neuronal co-cultures. Furthermore, Ad-IRF3 suppressed the expression of microRNA-155 and its star-form partner miR-155*, immunoregulatory miRNAs highly expressed in multiple sclerosis lesions. Astrocyte miR-155/miR155* were induced by cytokines and TLR ligands with a distinct hierarchy and involved in proinflammatory cytokine gene induction by targeting suppressor of cytokine signaling 1, a negative regulator of cytokine signaling and potentially other factors. Our results demonstrate a novel proinflammatory role for miR-155/miR-155* in human astrocytes and suggest that IRF3 can suppress neuroinflammation through regulating immunomodulatory miRNA expression. © 2011 Wiley-Liss, Inc.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Regulación de la Expresión Génica/genética , Factor 3 Regulador del Interferón/fisiología , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Técnicas de Cocultivo , Humanos , Factor 3 Regulador del Interferón/biosíntesis , Factor 3 Regulador del Interferón/genética , Fenotipo , Cultivo Primario de Células
8.
J Neuroinflammation ; 8: 187, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22208359

RESUMEN

BACKGROUND: Microglia are the principal cells involved in the innate immune response in the CNS. Activated microglia produce a number of proinflammatory cytokines implicated in neurotoxicity but they also are a major source of anti-inflammatory cytokines, antiviral proteins and growth factors. Therefore, an immune therapy aiming at suppressing the proinflammatory phenotype while enhancing the anti-inflammatory, growth promoting phenotype would be of great benefit. In the current study, we tested the hypothesis that interferon regulatory factor 3 (IRF3), a transcription factor required for the induction of IFNß following TLR3 or TLR4 activation, is critical to the microglial phenotype change from proinflammatory to anti-inflammatory, and that this phenotype change can be greatly facilitated by IRF3 gene transfer. METHODS: Cultures of primary human fetal microglia were transduced with IRF3 using recombinant adenovirus (Ad-IRF3) and subjected to microarray analysis, real-time PCR, immunoblotting and ELISA to determine inflammatory gene expression. Two different types of immune stimuli were tested, the TLR ligands, poly IC (PIC) and LPS, and the proinflammatory cytokines, IL-1/IFNγ. In addition, the role of the PI3K/Akt pathway was examined by use of a pharmacological inhibitor, LY294002. RESULTS: Our results show that Ad-IRF3 suppressed proinflammatory genes (IL-1α, IL-1ß, TNFα, IL-6, IL-8 and CXCL1) and enhanced anti-inflammatory genes (IL-1 receptor antagonist, IL-10 and IFNß) in microglia, regardless of the cell stimuli applied. Furthermore, Ad-IRF3 activated Akt, and LY294002 reversed the effects of Ad-IRF3 on microglial inflammatory gene expression. pAkt was critical in LPS- or PIC-induced production of IL-10 and IL-1ra. Significantly, microglial IFNß protein production was also dependent on pAkt and required both Ad-IRF3 and immunological stimuli (PIC > IL-1/IFNγ). pAkt played much less prominent and variable roles in microglial proinflammatory gene expression. This anti-inflammatory promoting role of PI3K/Akt appeared to be specific to microglia, since astrocyte proinflammatory gene expression (as well as IFNß expression) required PI3K/Akt. CONCLUSIONS: Our results show a novel anti-inflammatory role for the PI3K/Akt signaling pathway in microglia. They further suggest that IRF3 gene therapy could facilitate the microglial phenotype switch from proinflammatory ("M1-like") to anti-inflammatory and immunomodulatory ("M2-like"), in part, by augmenting the level of pAkt.


Asunto(s)
Factor 3 Regulador del Interferón/inmunología , Microglía/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/inmunología , Células Cultivadas , Quimiocinas/genética , Quimiocinas/inmunología , Cromonas/farmacología , Citocinas/genética , Citocinas/inmunología , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/genética , Interferón gamma/farmacología , Interleucina-1/farmacología , Análisis por Micromatrices , Microglía/citología , Microglía/efectos de los fármacos , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transgenes
9.
Contemp Clin Trials Commun ; 18: 100573, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32617430

RESUMEN

Crohn's disease (CD), a type of inflammatory bowel disease (IBD), is a chronic condition of the gastrointestinal tract that is caused by the loss of mucosal tolerance towards the commensal bacteria resulting in inflammatory responses. It has long been postulated that the gut microbiota, a complex and dynamic population of microorganisms, plays a key role in the pathogenesis of IBD. Maternal diagnosis of IBD has been identified as the greatest risk factor for IBD in offspring increasing the odds of developing the disease >4.5-fold. Moreover, babies born to mothers with IBD have demonstrated reduced gut bacterial diversity. There is accumulating evidence that the early life microbiota colonization is informed by maternal diet within the 3rd trimester of pregnancy. While babies born to mothers with IBD would pose an ideal cohort for intervention, no primary prevention measures are currently available. Therefore, we designed the MELODY (Modulating Early Life Microbiome through Dietary Intervention in Pregnancy) trial to test whether the IBD-AID™ dietary intervention during the last trimester of pregnancy can beneficially shift the microbiome of CD patients and their babies, thereby promoting a strong, effective immune system during a critical time of the immune system development. We will also test if favorable changes in the microbiome can lead to a reduced risk of postpartum CD relapse and lower mucosal inflammation in the offspring. This study will help create new opportunities to foster a healthy microbiome in the offspring at high risk of other immune-mediated diseases, potentially reducing their risk later in life.

10.
J Neurosci ; 26(14): 3845-54, 2006 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-16597739

RESUMEN

gamma-Secretase processing of the amyloid precursor protein (APP) generates Abeta40 and Abeta42, peptides that constitute the principal components of the beta-amyloid plaque pathology of Alzheimer's disease (AD). The gamma-secretase activity is executed by a high-molecular-weight complex of which presenilin 1 (PS1) is an essential component. PS1 is a multi-pass membrane protein, and the large hydrophilic loop domain between transmembrane domains 6 and 7 has been shown to interact with various proteins. To determine the physiological function of the loop domain, we created a strain of PS1 knock-in mice in which the exon 10, which encodes most of the hydrophilic loop sequence, was deleted from the endogenous PS1 gene. We report here that the homozygous exon 10-deleted mice are viable but exhibit drastically reduced gamma-secretase cleavage at the Abeta40, but not the Abeta42, site. Surprisingly, this reduction of Abeta40 is associated with exacerbated plaque pathology when expressed on APP transgenic background. Thus, the PS1 loop plays a regulatory role in gamma-secretase processing, and decreased Abeta40, not increased Abeta42 is likely the cause for the accelerated plaque deposition in these animals. Our finding supports a protective role of Abeta40 against amyloid pathology and raises the possibility that impaired gamma-secretase activity could be the basis for AD pathogenesis in general.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas , Activación Enzimática , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Presenilina-1 , Estructura Terciaria de Proteína , Relación Estructura-Actividad
11.
PLoS One ; 9(7): e103432, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25054228

RESUMEN

OBJECTIVE: Glioblastoma is the most frequent and malignant form of primary brain tumor with grave prognosis. Mounting evidence supports that chronic inflammation (such as chronic overactivation of IL-1 system) is a crucial event in carcinogenesis and tumor progression. IL-1 also is an important cytokine with species-dependent regulations and roles in CNS cell activation. While much attention is paid to specific anti-tumor immunity, little is known about the role of chronic inflammation/innate immunity in glioma pathogenesis. In this study, we examined whether human astrocytic cells (including malignant gliomas) can produce IL-1 and its role in glioma progression. METHODS: We used a combination of cell culture, real-time PCR, ELISA, western blot, immunocytochemistry, siRNA and plasmid transfection, micro-RNA analysis, angiogenesis (tube formation) assay, and neurotoxicity assay. RESULTS: Glioblastoma cells produced large quantities of IL-1 when activated, resembling macrophages/microglia. The activation signal was provided by IL-1 but not the pathogenic components LPS or poly IC. Glioblastoma cells were highly sensitive to IL-1 stimulation, suggesting its relevance in vivo. In human astrocytes, IL-1ß mRNA was not translated to protein. Plasmid transfection also failed to produce IL-1 protein, suggesting active repression. Suppression of microRNAs that can target IL-1α/ß did not induce IL-1 protein. Glioblastoma IL-1ß processing occurred by the NLRP3 inflammasome, and ATP and nigericin increased IL-1ß processing by upregulating NLRP3 expression, similar to macrophages. RNAi of annexin A2, a protein strongly implicated in glioma progression, prevented IL-1 induction, demonstrating its new role in innate immune activation. IL-1 also activated Stat3, a transcription factor crucial in glioma progression. IL-1 activated glioblastoma-conditioned media enhanced angiogenesis and neurotoxicity. CONCLUSIONS: Our results demonstrate unique, species-dependent immune activation mechanisms involving human astrocytes and astrogliomas. Specifically, the ability to produce IL-1 by glioblastoma cells may confer them a mesenchymal phenotype including increased migratory capacity, unique gene signature and proinflammatory signaling.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Anciano , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Células Cultivadas , Niño , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/inmunología , Glioma/patología , Humanos , Interleucina-1beta/inmunología , Masculino , Persona de Mediana Edad , ARN Mensajero/genética
12.
J Proteomics ; 99: 152-168, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24503185

RESUMEN

The tumor microenvironment including glial cells and their inflammatory products regulates brain tumor development and progression. We have previously established that human glioma cells are exquisitely sensitive to IL-1 stimulation leading us to undertake a comparative analysis of the secretome of unstimulated and cytokine (IL-1)-stimulated glioblastoma cells. We performed label-free quantitative proteomic analysis and detected 190 proteins which included cytokines, chemokines, growth factors, proteases, cell adhesion molecules, extracellular matrix (ECM) and related proteins. Measuring area under the curve (AUC) of peptides for quantitation, the IL-1-induced secretome contained 13 upregulated and 5 downregulated extracellular proteins (p<0.05) compared to controls. Of these, IL-8, CCL2, TNC, Gal-1 and PTX3 were validated as upregulated and SERPINE1, STC2, CTGF and COL4A2 were validated as downregulated factors by immunochemical methods. A major representation of the ECM and related proteins in the glioblastoma secretome and their modulation by IL-1 suggested that IL-1 induces its effect in part by altering TGFß expression, activity and signaling. These findings enhance our understanding of IL-1-induced modulation of glioma microenvironment, with implications for increased tumor invasion, migration and angiogenesis. They further provide novel targets for the glioblastoma intervention. BIOLOGICAL SIGNIFICANCE: Present study is on an unbiased screening of the glioblastoma secretome stimulated by IL-1 which triggers neuroinflammatory cascades in the central nervous system. Network of secreted proteins were shown to be regulated revealing their possible contribution to glioma progression. Label free quantitative proteomics has provided unique novel targets for potential glioblastoma intervention.


Asunto(s)
Movimiento Celular , Glioblastoma/metabolismo , Interleucina-1/farmacología , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteoma/metabolismo , Microambiente Tumoral , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Invasividad Neoplásica/patología , Neovascularización Patológica/patología
13.
J Neuroimmune Pharmacol ; 8(1): 132-44, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22684309

RESUMEN

Interferon regulatory factor 3 (IRF3) is a transcription factor critical in the induction of antiviral immunity. IRF3 is activated following stimulation of cell membrane or cytosolic nucleic acid sensors and is essential in the induction of the IFNß gene. Most cells constitutively express IRF3 in vitro, but little is known about the regulation of expression of IRF3 in vivo. Immunohistochemical analysis of selected human and mouse tissues demonstrated that IRF3 expression is highly organ- and cell-type specific, showing high expression in certain epithelial cells. In the CNS, while ependymal cells are strongly positive, brain parenchyma has little detectable IRF3 immunoreactivity. The importance of IRF3 in antiviral immunity has been demonstrated by the requirement for IRF3 in suppressing viral replication, but also by the demonstration that virus degrades IRF3 protein in infected cells. Furthermore, HIV-infected microglia in human CNS show abnormal IRF3+ aggregates, indicative of aberrant protein processing in vivo. In addition to antiviral immunity, IRF3 also plays a critical role in the modulation of neuroinflammation. A combination of dominant-negative and over-expression strategies in vitro as well as transgenic expression of IRF3 in vivo demonstrated that IRF3 plays a major role in modulating glial cytokine expression, i.e., suppression of proinflammatory cytokines and promotion of anti-inflammatory or immunoregulatory cytokines. These observations together suggest that IRF3 is a crucial regulator of immune responses against pathogen- and damage-associated molecules. We review recent literature on the molecular pathways of IRF3 activation and function of IRF3 and discuss their implications for CNS diseases.


Asunto(s)
Antiinflamatorios no Esteroideos , Antivirales , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Enfermedades del Sistema Nervioso Central/inmunología , Factor 3 Regulador del Interferón/farmacología , Animales , Enfermedades del Sistema Nervioso Central/prevención & control , Encefalitis/inmunología , Encefalitis/patología , Humanos , Factor 3 Regulador del Interferón/química
14.
PLoS One ; 8(6): e66657, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23885286

RESUMEN

Pannexin1 (Panx1) is a plasma membrane channel permeable to relatively large molecules, such as ATP. In the central nervous system (CNS) Panx1 is found in neurons and glia and in the immune system in macrophages and T-cells. We tested the hypothesis that Panx1-mediated ATP release contributes to expression of Experimental Autoimmune Encephalomyelitis (EAE), an animal model for multiple sclerosis, using wild-type (WT) and Panx1 knockout (KO) mice. Panx1 KO mice displayed a delayed onset of clinical signs of EAE and decreased mortality compared to WT mice, but developed as severe symptoms as the surviving WT mice. Spinal cord inflammatory lesions were also reduced in Panx1 KO EAE mice during acute disease. Additionally, pharmacologic inhibition of Panx1 channels with mefloquine (MFQ) reduced severity of acute and chronic EAE when administered before or after onset of clinical signs. ATP release and YoPro uptake were significantly increased in WT mice with EAE as compared to WT non-EAE and reduced in tissues of EAE Panx1 KO mice. Interestingly, we found that the P2X7 receptor was upregulated in the chronic phase of EAE in both WT and Panx1 KO spinal cords. Such increase in receptor expression is likely to counterbalance the decrease in ATP release recorded from Panx1 KO mice and thus contribute to the development of EAE symptoms in these mice. The present study shows that a Panx1 dependent mechanism (ATP release and/or inflammasome activation) contributes to disease progression, and that inhibition of Panx1 using pharmacology or gene disruption delays and attenuates clinical signs of EAE.


Asunto(s)
Conexinas/fisiología , Encefalomielitis Autoinmune Experimental/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Animales , Secuencia de Bases , Conexinas/genética , Cartilla de ADN , Encefalomielitis Autoinmune Experimental/metabolismo , Ensayo de Inmunoadsorción Enzimática , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Purinérgicos P2X7/metabolismo , Bazo/metabolismo , Bazo/patología
15.
PLoS One ; 7(4): e35115, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22509390

RESUMEN

BACKGROUND: The essential role of progranulin (PGRN) as a neurotrophic factor has been demonstrated by the discovery that haploinsufficiency due to GRN gene mutations causes frontotemporal lobar dementia. In addition to neurons, microglia in vivo express PGRN, but little is known about the regulation of PGRN expression by microglia. GOAL: In the current study, we examined the regulation of expression and function of PGRN, its proteolytic enzyme macrophage elastase (MMP-12), as well as the inhibitor of PGRN proteolysis, secretory leukocyte protease inhibitor (SLPI), in human CNS cells. METHODS: Cultures of primary human microglia and astrocytes were stimulated with the TLR ligands (LPS or poly IC), Th1 cytokines (IL-1/IFNγ), or Th2 cytokines (IL-4, IL-13). Results were analyzed by Q-PCR, immunoblotting or ELISA. The roles of MMP-12 and SLPI in PGRN cleavage were also examined. RESULTS: Unstimulated microglia produced nanogram levels of PGRN, and PGRN release from microglia was suppressed by the TLR ligands or IL-1/IFNγ, but increased by IL-4 or IL-13. Unexpectedly, while astrocytes stimulated with proinflammatory factors released large amounts of SLPI, none were detected in microglial cultures. We also identified MMP-12 as a PGRN proteolytic enzyme, and SLPI as an inhibitor of MMP-12-induced PGRN proteolysis. Experiments employing PGRN siRNA demonstrated that microglial PGRN was involved in the cytokine and chemokine production following TLR3/4 activation, with its effect on TNFα being the most conspicuous. CONCLUSIONS: Our study is the first detailed examination of PGRN in human microglia. Our results establish microglia as a significant source of PGRN, and MMP-12 and SLPI as modulators of PGRN proteolysis. Negative and positive regulation of microglial PGRN release by the proinflammatory/Th1 and the Th2 stimuli, respectively, suggests a fundamentally different aspect of PGRN regulation compared to other known microglial activation products. Microglial PGRN appears to function as an endogenous modulator of innate immune responses.


Asunto(s)
Astrocitos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Microglía , Astrocitos/enzimología , Astrocitos/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Metaloproteinasa 12 de la Matriz/genética , Microglía/enzimología , Microglía/metabolismo , Progranulinas , Proteolisis , ARN Interferente Pequeño , Inhibidor Secretorio de Peptidasas Leucocitarias/metabolismo , Transducción de Señal
16.
J Biol Chem ; 284(5): 2967-2977, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19036728

RESUMEN

gamma-Secretase is known to play a pivotal role in the pathogenesis of Alzheimer disease through production of amyloidogenic Abeta42 peptides. Early onset familial Alzheimer disease mutations in presenilin (PS), the catalytic core of gamma-secretase, invariably increase the Abeta42:Abeta40 ratio. However, the mechanism by which these mutations affect gamma-secretase complex formation and cleavage specificity is poorly understood. We show that our in vitro assay system recapitulates the effect of PS1 mutations on the Abeta42:Abeta40 ratio observed in cell and animal models. We have developed a series of small molecule affinity probes that allow us to characterize active gamma-secretase complexes. Furthermore we reveal that the equilibrium of PS1- and PS2-containing active complexes is dynamic and altered by overexpression of Pen2 or PS1 mutants and that formation of PS2 complexes is positively correlated with increased Abeta42:Abeta40 ratios. These data suggest that perturbations to gamma-secretase complex equilibrium can have a profound effect on enzyme activity and that increased PS2 complexes along with mutated PS1 complexes contribute to an increased Abeta42:Abeta40 ratio.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/fisiología , Proteínas de la Membrana/fisiología , Presenilina-1/fisiología , Presenilina-2/fisiología , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Biotina/metabolismo , Línea Celular , Flavina-Adenina Dinucleótido/genética , Flavina-Adenina Dinucleótido/fisiología , Técnicas de Sustitución del Gen , Células HeLa , Humanos , Ratones , Presenilina-1/metabolismo , Presenilina-2/metabolismo
17.
Chemistry ; 14(14): 4293-306, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18366032

RESUMEN

Deoxyharringtonine (2), homoharringtonine (3), homodeoxyharringtonine (4), and anhydroharringtonine (5) are reported to be among the most potent members of the antileukemia alkaloids isolated from the Cephalotaxus genus. Convergent syntheses of these four natural products are described, each involving novel synthetic methods and strategies. These syntheses enabled evaluation of several advanced natural and non-natural compounds against an array of human hematopoietic and solid tumor cells. Potent cytotoxicity was observed in several cell lines previously not challenged with these alkaloids. Variations in the structure of the ester chain within this family of alkaloids confer differing activity profiles against vincristine-resistant HL-60/RV+, signalling new avenues for molecular design of these natural products to combat multi-drug resistance.


Asunto(s)
Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/farmacología , Harringtoninas/síntesis química , Harringtoninas/farmacología , Aziridinas/química , Benzazepinas/síntesis química , Benzazepinas/química , Línea Celular Tumoral , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Ésteres/síntesis química , Ésteres/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Homoharringtonina , Humanos , Retinoblastoma/tratamiento farmacológico
18.
J Org Chem ; 69(21): 7344-7, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15471490

RESUMEN

The first asymmetric synthesis of novel, potent photoreactive gamma-secretase inhibitors 2 and 3 has been accomplished. Two stereoselective methods for the preparation of lactone 9 are described. Protected benzophenone intermediate 19 is prepared via an aldol-elimination reaction followed by a PtO(2)-catalyzed asymmetric hydrogenation. Two routes leading from 19 to compounds 2 and 3 are evaluated. The application of 3 as an activity-based probe has been demonstrated by localizing gamma-secretase activity in the plasma membrane of intact cells.


Asunto(s)
Endopeptidasas/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Fragmentos de Péptidos/química , Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Carbamatos/química , Carbamatos/farmacología , Membrana Celular/química , Membrana Celular/enzimología , Dipéptidos/química , Dipéptidos/farmacología , Endopeptidasas/química , Inhibidores Enzimáticos/farmacología , Humanos , Conformación Molecular , Imitación Molecular , Fotoquímica , Estereoisomerismo
19.
Proc Natl Acad Sci U S A ; 101(49): 17050-5, 2004 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-15563588

RESUMEN

gamma-Secretase activity is associated with a presenilin (PS)-containing macromolecular complex. Whether PS contains the active site of gamma-secretase has been controversial. One challenge is to find PS that is engaged in the active gamma-secretase complex at the cell surface, where some substrates appear to be processed. In this study, we developed an intact cell photolabeling technique that allows the direct visualization of active gamma-secretase at the cell surface. We demonstrated that active gamma-secretase is present in the plasma membrane. Moreover, the PS1 heterodimer is specifically photolabeled at the cell surface by a potent inhibitor that binds to only the active gamma-secretase. We also explored the cellular processing sites of gamma-secretase for amyloid precursor protein (APP) and Notch by using small molecular probes. MRL631, a gamma-secretase inhibitor that is unable to penetrate the cell membrane, significantly blocks gamma-secretase-mediated Notch cleavage but has little effect on APP processing. These results indicate that Notch is processed at the cell surface and that the majority of APP is processed by intracellular gamma-secretase. Furthermore, the fact that inhibitors first target gamma-secretase in the plasma membrane for Notch processing, and not for APP, will have important implications for drug development to treat Alzheimer's disease and cancer.


Asunto(s)
Amiloide/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Línea Celular , Membrana Celular , Humanos , Proteínas de la Membrana/genética , Etiquetas de Fotoafinidad , Inhibidores de Proteasas/farmacología , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Receptores Notch , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA