Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
PLoS Genet ; 19(1): e1010586, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36622851

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is one of the most frequent causes of pneumonia, sepsis and meningitis in humans, and an important cause of mortality among children and the elderly. We have previously reported the suitability of the zebrafish (Danio rerio) larval model for the study of the host-pathogen interactions in pneumococcal infection. In the present study, we characterized the zebrafish innate immune response to pneumococcus in detail through a whole-genome level transcriptome analysis and revealed a well-conserved response to this human pathogen in challenged larvae. In addition, to gain understanding of the genetic factors associated with the increased risk for severe pneumococcal infection in humans, we carried out a medium-scale forward genetic screen in zebrafish. In the screen, we identified a mutant fish line which showed compromised resistance to pneumococcus in the septic larval infection model. The transcriptome analysis of the mutant zebrafish larvae revealed deficient expression of a gene homologous for human C-reactive protein (CRP). Furthermore, knockout of one of the six zebrafish crp genes by CRISPR-Cas9 mutagenesis predisposed zebrafish larvae to a more severe pneumococcal infection, and the phenotype was further augmented by concomitant knockdown of a gene for another Crp isoform. This suggests a conserved function of C-reactive protein in anti-pneumococcal immunity in zebrafish. Altogether, this study highlights the similarity of the host response to pneumococcus in zebrafish and humans, gives evidence of the conserved role of C-reactive protein in the defense against pneumococcus, and suggests novel host genes associated with pneumococcal infection.


Subject(s)
Pneumococcal Infections , Zebrafish , Animals , Child , Humans , Aged , Zebrafish/genetics , C-Reactive Protein , Pneumococcal Infections/genetics , Immunity, Innate/genetics , Streptococcus pneumoniae/genetics
2.
Pathology ; 53(7): 875-882, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34049715

ABSTRACT

B-cell lineage acute lymphoblastic leukaemia (B-ALL) is the most common paediatric malignancy. Transcription factor B-cell lymphoma 6 (BCL6) is essential to germinal centre formation and antibody affinity maturation and plays a major role in mature B-cell malignancies. More recently, it was shown to act as a critical downstream regulator in pre-BCR+ B-ALL. We investigated the expression of the BCL6 protein in a population-based cohort of paediatric B-ALL cases and detected moderate to strong positivity through immunohistochemistry in 7% of cases (8/117); however, only two of eight BCL6 cases (25%) co-expressed the ZAP70 protein. In light of these data, the subtype with active pre-BCR signalling constitutes a rare entity in paediatric B-ALL. In three independent larger cohorts with gene expression data, high BCL6 mRNA levels were associated with the TCF3-PBX1, Ph-like, NUTM1, MEF2D and PAX5-alt subgroups and the 'metagene' signature for pre-BCR-associated genes. However, higher-than-median BCL6 mRNA level alone was associated with favourable event free survival in the Nordic paediatric cohort, indicating that using BCL6 as a diagnostic marker requires careful design, and evaluation of protein level is needed alongside the genetic or transcriptomic data.


Subject(s)
DNA Copy Number Variations , Lymphoma, B-Cell/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Proto-Oncogene Proteins c-bcl-6/metabolism , B-Lymphocytes/pathology , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Pediatrics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Progression-Free Survival , Proto-Oncogene Proteins c-bcl-6/genetics , RNA, Messenger/genetics
3.
Genome Med ; 12(1): 99, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33218352

ABSTRACT

BACKGROUND: Tight regulatory loops orchestrate commitment to B cell fate within bone marrow. Genetic lesions in this gene regulatory network underlie the emergence of the most common childhood cancer, acute lymphoblastic leukemia (ALL). The initial genetic hits, including the common translocation that fuses ETV6 and RUNX1 genes, lead to arrested cell differentiation. Here, we aimed to characterize transcription factor activities along the B-lineage differentiation trajectory as a reference to characterize the aberrant cell states present in leukemic bone marrow, and to identify those transcription factors that maintain cancer-specific cell states for more precise therapeutic intervention. METHODS: We compared normal B-lineage differentiation and in vivo leukemic cell states using single cell RNA-sequencing (scRNA-seq) and several complementary genomics profiles. Based on statistical tools for scRNA-seq, we benchmarked a workflow to resolve transcription factor activities and gene expression distribution changes in healthy bone marrow lymphoid cell states. We compared these to ALL bone marrow at diagnosis and in vivo during chemotherapy, focusing on leukemias carrying the ETV6-RUNX1 fusion. RESULTS: We show that lymphoid cell transcription factor activities uncovered from bone marrow scRNA-seq have high correspondence with independent ATAC- and ChIP-seq data. Using this comprehensive reference for regulatory factors coordinating B-lineage differentiation, our analysis of ETV6-RUNX1-positive ALL cases revealed elevated activity of multiple ETS-transcription factors in leukemic cells states, including the leukemia genome-wide association study hit ELK3. The accompanying gene expression changes associated with natural killer cell inactivation and depletion in the leukemic immune microenvironment. Moreover, our results suggest that the abundance of G1 cell cycle state at diagnosis and lack of differentiation-associated regulatory network changes during induction chemotherapy represent features of chemoresistance. To target the leukemic regulatory program and thereby overcome treatment resistance, we show that inhibition of ETS-transcription factors reduced cell viability and resolved pathways contributing to this using scRNA-seq. CONCLUSIONS: Our data provide a detailed picture of the transcription factor activities characterizing both normal B-lineage differentiation and those acquired in leukemic bone marrow and provide a rational basis for new treatment strategies targeting the immune microenvironment and the active regulatory network in leukemia.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia/genetics , Lymphocytes/physiology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Bone Marrow , Cell Line, Tumor , Child , Core Binding Factor Alpha 2 Subunit/metabolism , Drug Delivery Systems , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Leukemia/drug therapy , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Transcription Factors , Transcriptome , Translocation, Genetic , ETS Translocation Variant 6 Protein
4.
Sci Rep ; 10(1): 2043, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029838

ABSTRACT

Acute lymphoblastic leukemia is marked by aberrant transcriptional features that alter cell differentiation, self-renewal, and proliferative features. We sought to identify the transcription factors exhibiting altered and subtype-specific expression patterns in B-ALL and report here that SOX11, a developmental and neuronal transcription factor, is aberrantly expressed in the ETV6-RUNX1 and TCF3-PBX1 subtypes of acute B-cell leukemias. We show that a high expression of SOX11 leads to alterations of gene expression that are typically associated with cell adhesion, migration, and differentiation. A high expression is associated with DNA hypomethylation at the SOX11 locus and a favorable outcome. The results indicate that SOX11 expression marks a group of patients with good outcomes and thereby prompts further study of its use as a biomarker.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , SOXC Transcription Factors/metabolism , Adolescent , Biopsy , Bone Marrow/pathology , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/genetics , DNA Methylation , Disease-Free Survival , Female , Follow-Up Studies , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Infant , Male , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , SOXC Transcription Factors/genetics
5.
J Allergy Clin Immunol ; 140(3): 782-796, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28115215

ABSTRACT

BACKGROUND: The nuclear factor κ light-chain enhancer of activated B cells (NF-κB) signaling pathway is a key regulator of immune responses. Accordingly, mutations in several NF-κB pathway genes cause immunodeficiency. OBJECTIVE: We sought to identify the cause of disease in 3 unrelated Finnish kindreds with variable symptoms of immunodeficiency and autoinflammation. METHODS: We applied genetic linkage analysis and next-generation sequencing and functional analyses of NFKB1 and its mutated alleles. RESULTS: In all affected subjects we detected novel heterozygous variants in NFKB1, encoding for p50/p105. Symptoms in variant carriers differed depending on the mutation. Patients harboring a p.I553M variant presented with antibody deficiency, infection susceptibility, and multiorgan autoimmunity. Patients with a p.H67R substitution had antibody deficiency and experienced autoinflammatory episodes, including aphthae, gastrointestinal disease, febrile attacks, and small-vessel vasculitis characteristic of Behçet disease. Patients with a p.R157X stop-gain experienced hyperinflammatory responses to surgery and showed enhanced inflammasome activation. In functional analyses the p.R157X variant caused proteasome-dependent degradation of both the truncated and wild-type proteins, leading to a dramatic loss of p50/p105. The p.H67R variant reduced nuclear entry of p50 and showed decreased transcriptional activity in luciferase reporter assays. The p.I553M mutation in turn showed no change in p50 function but exhibited reduced p105 phosphorylation and stability. Affinity purification mass spectrometry also demonstrated that both missense variants led to altered protein-protein interactions. CONCLUSION: Our findings broaden the scope of phenotypes caused by mutations in NFKB1 and suggest that a subset of autoinflammatory diseases, such as Behçet disease, can be caused by rare monogenic variants in genes of the NF-κB pathway.


Subject(s)
Autoimmune Diseases/genetics , Immunologic Deficiency Syndromes/genetics , NF-kappa B/genetics , Adult , Aged , Cell Line , Child , Female , Heterozygote , Humans , Inflammation/genetics , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Mutation , Phenotype
7.
N Engl J Med ; 372(25): 2409-22, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26083206

ABSTRACT

Background Combined immunodeficiencies are marked by inborn errors of T-cell immunity in which the T cells that are present are quantitatively or functionally deficient. Impaired humoral immunity is also common. Patients have severe infections, autoimmunity, or both. The specific molecular, cellular, and clinical features of many types of combined immunodeficiencies remain unknown. Methods We performed genetic and cellular immunologic studies involving five unrelated children with early-onset invasive bacterial and viral infections, lymphopenia, and defective T-cell, B-cell, and natural killer (NK)-cell responses. Two patients died early in childhood; after allogeneic hematopoietic stem-cell transplantation, the other three had normalization of T-cell function and clinical improvement. Results We identified biallelic mutations in the dedicator of cytokinesis 2 gene (DOCK2) in these five patients. RAC1 activation was impaired in the T cells. Chemokine-induced migration and actin polymerization were defective in the T cells, B cells, and NK cells. NK-cell degranulation was also affected. Interferon-α and interferon-λ production by peripheral-blood mononuclear cells was diminished after viral infection. Moreover, in DOCK2-deficient fibroblasts, viral replication was increased and virus-induced cell death was enhanced; these conditions were normalized by treatment with interferon alfa-2b or after expression of wild-type DOCK2. Conclusions Autosomal recessive DOCK2 deficiency is a new mendelian disorder with pleiotropic defects of hematopoietic and nonhematopoietic immunity. Children with clinical features of combined immunodeficiencies, especially with early-onset, invasive infections, may have this condition. (Supported by the National Institutes of Health and others.).


Subject(s)
Genetic Diseases, Inborn/genetics , Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , T-Lymphocytes/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Child, Preschool , Fatal Outcome , Female , GTPase-Activating Proteins , Genes, Recessive , Genetic Diseases, Inborn/therapy , Guanine Nucleotide Exchange Factors/deficiency , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Deficiency Syndromes/therapy , Infant , Killer Cells, Natural/immunology , Male , Pedigree , T-Lymphocytes/metabolism , rac1 GTP-Binding Protein/metabolism
8.
Dev Comp Immunol ; 42(2): 345-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24076065

ABSTRACT

Streptococcus pneumoniae (pneumococcus) is a leading cause of community acquired pneumonia, septicemia, and meningitis. Due to incomplete understanding of the host and bacterial factors contributing to these diseases optimal treatment and prevention methods are lacking. In the present study we examined whether the adult zebrafish (Danio rerio) can be used to investigate the pathophysiology of pneumococcal diseases. Here we show that both intraperitoneal and intramuscular injections of the pneumococcal strain TIGR4 cause a fulminant, dose-dependent infection in adult zebrafish, while isogenic mutant bacteria lacking the polysaccharide capsule, autolysin, or pneumolysin are attenuated in the model. Infection through the intraperitoneal route is characterized by rapid expansion of pneumococci in the bloodstream, followed by penetration of the blood-brain barrier and progression to meningitis. Using Rag1 mutant zebrafish, which are devoid of somatic recombination and thus lack adaptive immune responses, we show that clearance of pneumococci in adult zebrafish depends mainly on innate immune responses. In conclusion, this study provides evidence that the adult zebrafish can be used as a model for a pneumococcal infection, and that it can be used to study both host and bacterial factors involved in the pathogenesis. However, our results do not support the use of the zebrafish in studies on the role of adaptive immunity in pneumococcal disease or in the development of new pneumococcal vaccines.


Subject(s)
Disease Models, Animal , Meningitis, Bacterial/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae , Zebrafish/immunology , Animals , Animals, Genetically Modified , Bacterial Proteins/genetics , Homeodomain Proteins/genetics , Meningitis, Bacterial/physiopathology , Pneumococcal Infections/physiopathology , Polysaccharides, Bacterial , Streptolysins/genetics , Zebrafish/genetics , Zebrafish/microbiology
9.
Duodecim ; 129(9): 939-46, 2013.
Article in Finnish | MEDLINE | ID: mdl-23786106

ABSTRACT

Most cases of childhood leukemia are acute lymphoblastic leukemia, a small proportion being myelogenous leukemia. Chronic myelogenous leukemia is rare in children, whereas chronic lymphocytic leukemia is not encountered. The prognosis of childhood leukemia has improved considerably so that approximately 80 to 90% of those having lymphocytic leukemia and 60 to 70% of those having myelogenous leukemia will recover fully. The challenge is to develop new treatments for disease groups having a poor prognosis and on the other hand to lighten the treatments for leukemias having a good prognosis.


Subject(s)
Leukemia/pathology , Leukemia/therapy , Child , Humans , Prognosis
10.
mBio ; 4(1): e00535-12, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23269830

ABSTRACT

UNLABELLED: Pneumococcal pili have been shown to influence pneumococcal colonization, disease development, and the inflammatory response in mice. The role of the pilus-associated RrgA adhesin in pneumococcal interactions with murine and human macrophages was investigated. Expression of pili with RrgA enhanced the uptake of pneumococci by murine and human macrophages that was abolished by antibodies to complement receptor 3 (CR3) and not seen in CR3-deficient macrophages. Recombinant RrgA, but not pilus subunit RrgC, promoted CR3-mediated phagocytosis of coated beads by murine and human macrophages. Flow cytometry showed that purified CR3 binds pneumococcal cells expressing RrgA, and purified RrgA was shown to interact with CR3 and its I domain. In vivo, RrgA facilitated spread of pneumococci from the upper airways and peritoneal cavity to the bloodstream. Earlier onset of septicemia and more rapidly progressing disease was observed in wild-type mice compared to CR3-deficient mice challenged intranasally or intraperitoneally with pneumococci. Motility assays and time-lapse video microscopy showed that pneumococcal stimulation of macrophage motility required RrgA and CR3. These findings, together with the observed RrgA-dependent increase of intracellular survivors up to 10 h following macrophage infection, suggest that RrgA-CR3-mediated phagocytosis promotes systemic pneumococcal spread from local sites. IMPORTANCE: Streptococcus pneumoniae is a major contributor to morbidity and mortality in infectious diseases globally. Symptomatology is mainly due to pneumococcal interactions with host cells leading to an inflammatory response. However, we still need more knowledge on how pneumococci talk to immune cells and the importance of this interaction. Recently, a novel structure was identified on the pneumococcal surface, an adhesive pilus found in about 30% of clinical pneumococcal isolates. The pilus has been suggested to be important for successful spread of antibiotic-resistant pneumococcal clones globally. Here we sought to identify mechanisms for how the pneumococcal pilin subunit RrgA contributes to disease development by interacting with host immune cells. Our data suggest a new way for how pneumococci may cross talk with phagocytic cells and affect disease progression. An increased understanding of these processes may lead to better strategies for how to treat these common infections.


Subject(s)
Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Macrophages/immunology , Streptococcus pneumoniae/immunology , Virulence Factors/immunology , Virulence Factors/metabolism , Adhesins, Bacterial/immunology , Adhesins, Bacterial/metabolism , Animals , Blood/microbiology , Cell Movement , Female , Flow Cytometry , Humans , Macrophages/microbiology , Male , Mice , Peritoneal Cavity/microbiology , Phagocytosis , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Protein Binding , Protein Interaction Domains and Motifs , Respiratory System/microbiology , Streptococcus pneumoniae/pathogenicity , Survival Analysis , Time-Lapse Imaging
11.
Dev Comp Immunol ; 38(3): 447-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22867759

ABSTRACT

Streptococcus agalactiae (Group B Streptococcus, GBS) is the major cause of severe bacterial disease and meningitis in newborns. The zebrafish (Danio rerio) has recently emerged as a valuable and powerful vertebrate model for the study of human streptococcal infections. In the present study we demonstrate that adult zebrafish are susceptible to GBS infection through the intraperitoneal and intramuscular routes of infection. Following intraperitoneal challenge with GBS, zebrafish developed a fulminant infection 24-48 h post-injection, with signs of pathogenesis including severe inflammation at the injection site and meningoencephalitis. Quantification of blood and brain bacterial load confirmed that GBS is capable of replicating in the zebrafish bloodstream and penetrating the blood-brain barrier, resulting in the induction of host inflammatory immune responses in the brain. Additionally, we show that GBS mutants previously described as avirulent in the mice model, have an impaired ability to cause meningitis in this new in vivo model. Taken together, our data demonstrates that adult zebrafish may be used as a bacterial meningitis model as a means for deciphering the pathogenesis and development of invasive GBS disease.


Subject(s)
Disease Models, Animal , Meningitis, Bacterial , Streptococcal Infections , Streptococcus agalactiae , Zebrafish , Animals , Bacterial Load , Blood-Brain Barrier/microbiology , Brain/immunology , Brain/microbiology , Brain/pathology , Meningitis, Bacterial/immunology , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Meningoencephalitis/immunology , Meningoencephalitis/microbiology , Meningoencephalitis/pathology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity
12.
Dev Comp Immunol ; 36(2): 342-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21658407

ABSTRACT

Severe community acquired pneumonia caused by Streptococcus pneumoniae is the most common cause of death from infection in developing countries. Serotype specific conjugate vaccines have decreased the incidence of invasive infections, but at the same time, disease due to non-vaccine serotypes have increased. New insights into host immune mechanisms against pneumococcus may provide better treatment and prevention strategies. Zebrafish is an attractive vertebrate model for studying host immune responses and infection biology. Here we show that an intravenous challenge with pneumococcus infects zebrafish embryos leading to death in a dose dependent manner. Survival rates correlate with the bacterial burden in the embryos. The production of proinflammatory cytokines is induced in zebrafish after pneumococcal exposure. Importantly, morpholino treated embryos lacking either myeloid cells or the ability to phagocytose bacteria have lowered survival rates compared to wild type embryos after pneumococcal challenge. These data suggest that the survival of zebrafish embryos upon intravenous infection with S. pneumoniae is dependent on the clearance of the bacteria by phagocytosing cells. Additionally, we demonstrate that mutant pneumococci lacking known virulence factors are attenuated in the zebrafish model. Our data demonstrate that zebrafish embryos can be used for study innate immune responses as well as virulence determinants in pneumococcal infections.


Subject(s)
Host-Pathogen Interactions/immunology , Phagocytosis/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Embryo, Nonmammalian , Microscopy, Fluorescence , Mutagenesis, Insertional/methods , Pneumococcal Infections/microbiology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Virulence Factors/genetics , Virulence Factors/immunology , Zebrafish
13.
Cell Signal ; 20(10): 1873-81, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18652890

ABSTRACT

Both type I interferons (IFNs) and interferon regulatory factors (IRFs) are well characterized in viral infections, whereas they are far less studied in bacterially activated toll-like receptor (TLR) pathways. Here, we studied the involvement of IRF1 and IRF2 in TLR2-mediated responses. In mouse macrophages, IRF2 was activated by lipoteichoic acid (LTA) of Staphylococcus aureus, resulting in up-regulation of IRF1 and rapid secretion of IFN-alpha. In addition, LTA-induced activation of Signal transducers and activators of transcription 1 (Stat1) and Stat3 via IRF2. The secretion of IFN-alpha was reduced in IRF2-silenced macrophages, resulting in a disappearance of tyrosine-phosphorylated Stat3 and a reduction of pro-inflammatory responses, despite induction of Mal adapter protein. These results provide a mechanistic insight into the pro-inflammatory responses against S. aureus LTA in mouse macrophages. IRFs can be intersecting factors of viral and bacterial responses in activated TLR signalling pathways.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/metabolism , Interferon-alpha/biosynthesis , STAT Transcription Factors/metabolism , Signal Transduction , Staphylococcus aureus/chemistry , Toll-Like Receptor 2/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Cell Line , Inflammation , Interferon-alpha/metabolism , Interferon-beta/metabolism , Interferon-gamma/metabolism , Ligands , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Protein-Tyrosine Kinases/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Teichoic Acids/pharmacology , Transcription Factors/metabolism
14.
Pediatr Res ; 59(2): 180-4, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439575

ABSTRACT

We hypothesized that, in acute endotoxin-induced fetal cardiac dysfunction, atrial (ANP) and B-type (BNP) natriuretic peptide mRNA expressions are increased in proportion to the severity of fetal cardiovascular compromise in mouse. To investigate in vitro the effect of endotoxin-induced inflammation on cardiac natriuretic peptide expression, fetal hearts were harvested at 15-16 d of gestation and incubated for 6 h with lipopolysaccharide (LPS). To examine the relationship between fetal cardiovascular compromise and cardiac natriuretic peptide expression in endotoxin-induced cardiac dysfunction in the in vivo model, fetuses received intra-amniotically 25 microL LPS (10 microg/mL) or 25 microL of 0.9% saline. Fetal Doppler ultrasonography was performed before and six hours after the injections. In in vitro cultured fetal hearts, LPS induced the production of proinflammatory cytokines without affecting the basal expressions of natriuretic peptides. In the in vivo model, Doppler ultrasonography revealed severe cardiac dysfunction after LPS injection. No significant changes in ANP or atrial BNP mRNA were found. The fetal ventricular BNP mRNA levels were about 2.6-fold in the LPS group compared with the control group. Decreased fetal cardiac outflow mean velocity, increased proportion of isovolumetric contraction time of the cardiac cycle, and increased pulsatility indices of the descending aorta and inferior vena cava were related to elevated ventricular BNP mRNA levels. Our results show that LPS did not increase the mRNA expression of natriuretic peptides in cultured fetal hearts. In contrast, fetal ventricular BNP gene expression was increased in proportion to the severity of the hemodynamic compromise in vivo.


Subject(s)
Atrial Natriuretic Factor/genetics , Endotoxins/toxicity , Heart/physiopathology , Animals , Base Sequence , DNA Primers , Female , Heart/embryology , Hemodynamics , Mice , Mice, Inbred DBA , Pregnancy , RNA, Messenger/genetics
15.
Am J Pathol ; 166(6): 1585-92, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15920144

ABSTRACT

Intra-amniotic lipopolysaccharide (LPS) causes an acute inflammatory response and cardiac dysfunction in fetal mice. We hypothesized that the placenta protects the fetus against maternally administered bacterial toxins, delaying the onset of a fetal inflammatory response and vascular compromise. At 14 to 15 days of gestation, DBA mice were randomized to receive LPS (2.4 mg/kg) or vehicle intraperitoneally. Doppler ultrasonography of fetal cardiovascular hemodynamics was performed before and 6 hours after maternal LPS. Six hours after the LPS, maternal serum concentrations of tumor necrosis factor-alpha and interleukin (IL)-6 (P < 0.05) were increased. Placenta showed severe maternal vascular dilatation and congestion. The expressions of tumor necrosis factor-alpha, IL-1alpha, and IL-6 (P < 0.05) were increased, and the expression of Toll-like receptor 4 was constitutive in placenta. The expression of Toll-like receptor 2 increased (P < 0.05) and was detected in labyrinthine macrophages. No inflammatory activation was found in fetal tissues, and amniotic fluid revealed no significant increase in cytokines. The ultrasonographic examination demonstrated increased fetal cardiac afterload after LPS, with 65% of the fetuses exhibiting atrioventricular valve regurgitation. In conclusion, maternal inflammatory insult activates placental labyrinthine macrophages leading to an acute increase in placental vascular resistance and fetal cardiac dysfunction without an inflammatory response in fetus.


Subject(s)
Cardiovascular System/drug effects , Fetal Diseases/immunology , Inflammation/pathology , Placenta/pathology , Pregnancy Complications/physiopathology , Amniotic Fluid/chemistry , Amniotic Fluid/drug effects , Amniotic Fluid/immunology , Animals , Cytokines/analysis , Cytokines/genetics , Echocardiography, Doppler , Female , Fetal Diseases/etiology , Fetal Diseases/pathology , Fetus , Gene Expression , Inflammation/chemically induced , Inflammation/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/immunology , Macrophages/immunology , Mice , Placenta/immunology , Placenta/metabolism , Pregnancy , RNA, Messenger/analysis
16.
Pediatr Res ; 57(5 Pt 1): 644-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15718365

ABSTRACT

Endotoxin [lipopolysaccharide (LPS)] from Gram-negative bacteria is found in amniotic fluid in intrauterine infections that associate with the risk for spontaneous premature birth, bronchopulmonary dysplasia (BPD), and respiratory distress syndrome. Toll-like receptor 4 (TLR4) is the signaling receptor for LPS. The aim was to investigate the primary inflammatory response in mice shortly after administration of LPS to the dam (14 and 17 d of pregnancy), to the newborn, or into the amniotic fluid. The expression levels of TLR4, IL-1, tumor necrosis factor-alpha, IL-6, IL-10, macrophage inflammatory protein-2, and IL-1 receptor 1 were studied with ribonuclease protection assay. In addition, TLR4 protein was analyzed with Western blotting. The fetal membranes expressed TLR4 mRNA and protein and showed an acute cytokine response to LPS when LPS was administrated into the amniotic fluid. There was distinct ontogeny in the responsiveness of fetal lung to LPS: on fetal day 14 (term 20 d), both the expression of TLR4 and the acute cytokine response were undetectable 5 h after LPS; they became detectable by fetal day 17. TLR4 and the cytokine response further increased after birth. In maternal lung, the TLR4 expression was strongest and up-regulated in parallel with the induction of the cytokines. We propose that TLR4 controls the magnitude of the LPS-induced cytokine response during the perinatal period.


Subject(s)
Endotoxins/metabolism , Membrane Glycoproteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Alleles , Animals , Animals, Newborn , Blotting, Western , Chemokine CXCL2 , Cytokines/metabolism , Disease Models, Animal , Inflammation , Interleukin-1/biosynthesis , Interleukin-10/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/metabolism , Lung Diseases/microbiology , Mice , Mice, Inbred DBA , Monokines/biosynthesis , Placenta/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Interleukin-1/biosynthesis , Receptors, Interleukin-1 Type I , Ribonucleases/metabolism , Signal Transduction , Time Factors , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
17.
Biol Neonate ; 87(3): 152-9, 2005.
Article in English | MEDLINE | ID: mdl-15564781

ABSTRACT

Interleukin-1 (IL-1) increases the expression of surfactant protein A (SP-A) in rabbit, lamb and human fetal lung. The upregulation disappears towards term. Among the transcription factors, IL-1 activates nuclear factor kappaB (NF-kappaB) and CCAAT/enhancer-binding protein delta (C/EBPdelta). NF-kappaB presumably has a role in IL-1-induced upregulation of SP-A. Also, C/EBPdelta may regulate SP-A expression. The aim was to study the role of these transcription factors in the induced effect of IL-1 on SP-A expression. Explants from fetal and neonatal rabbit lung were cultured in vitro followed by studies using immunohistochemistry, electrophoretic mobility shift assay and Northern analysis. We found gestation-dependent changes in IL-1-induced immunoreactivities of NF-kappaB and C/EBPdelta in the nuclei of alveolar cells. This increase in nuclear transcription factors correlated with IL-1-induced SP-A expression levels. As studied in the explants from fetal and newborn lung, the SP-A mRNA expression additionally associated with C/EBPdelta mRNA and with the binding of nuclear extracts from the lung explants to the C/EBP consensus probe. On the basis of the present and previous studies, we propose that NF-kappaB and C/EBPdelta have potential mediator roles in IL-1-induced upregulation of SP-A in immature lung.


Subject(s)
Animals, Newborn/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Interleukin-1/pharmacology , Lung/embryology , Lung/metabolism , NF-kappa B/physiology , Pulmonary Surfactant-Associated Protein A/metabolism , Recombinant Proteins/pharmacology , Transcription Factors/physiology , Animals , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Consensus Sequence , Fetus/metabolism , Humans , Immunohistochemistry , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Organ Culture Techniques , Pulmonary Surfactant-Associated Protein A/genetics , RNA, Messenger/metabolism , Rabbits , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Cardiovasc Res ; 60(1): 156-64, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14522418

ABSTRACT

OBJECTIVE: Intrauterine infection is associated with increased lipopolysaccharide (LPS) and proinflammatory cytokines in amniotic fluid. We hypothesized that intra-amniotic LPS launches a fetal inflammatory response leading to cardiac dysfunction. METHODS: A mouse model was established. At 15-16 days of gestation, 52 fetuses of nine dams received LPS and 46 fetuses of nine dams vehicle intra-amniotically. Five dams underwent a sham operation. Echocardiography was performed before and 6 h after the injection to obtain inflow and outflow blood velocity waveforms. Outflow mean velocity (V(mean)) and the proportions of isovolumetric relaxation (IRT%) and contraction (ICT%) times of the cardiac cycle were calculated. Pulsatility indices (PI) were calculated from the umbilical and intracranial arteries and the descending aorta. Pulsatility indices for veins (PIV) were obtained from ductus venosus. Toll-like receptor-4 (TLR4) and several other inflammatory mediators were determined using ELISA, immunohistochemistry, or ribonuclease protection assay. RESULTS: In the LPS group, outflow V(mean) was significantly lower, and ICT% and IRT% longer than in the other groups. LPS increased PIs, except in the intracranial arteries, which showed a decrease in PIs. In ductus venosus, PIVs were increased after LPS. LPS increased interleukin (IL)-6 in amniotic fluid and induced the expression of proinflammatory cytokines in placenta and fetal membranes, but not in lung. In fetal myocardium, TLR4 was constitutional. LPS induced the expression of IL-1beta and tumor necrosis factor (TNF)-alpha mRNA in myocardium, whereas inducible nitric oxide synthase (NOS2) protein and nitrotyrosine remained undetectable. CONCLUSIONS: As a response to endotoxin in amniotic fluid, fetal myocardium acutely generates cytokines and severe fetal cardiovascular compromise develops. These two may be linked through a mechanism that does not include NO.


Subject(s)
Amniotic Fluid/immunology , Fetal Diseases/immunology , Myocardial Contraction , Systemic Inflammatory Response Syndrome/embryology , Systemic Inflammatory Response Syndrome/immunology , Animals , Blood Flow Velocity , Dose-Response Relationship, Drug , Echocardiography, Doppler , Extraembryonic Membranes/immunology , Female , Interleukin-6/analysis , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/analysis , Mice , Mice, Inbred DBA , Models, Animal , Placenta/immunology , Pregnancy , Receptors, Cell Surface/analysis , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/analysis , Ultrasonography, Prenatal
SELECTION OF CITATIONS
SEARCH DETAIL
...