Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 10: 355, 2019.
Article in English | MEDLINE | ID: mdl-30886604

ABSTRACT

Molecular determinants underlying interferon (IFN)-macrophage biology can help delineate enzyme systems, pathways and mechanisms for enabling host-directed therapeutic approaches against infection. Notably, while the IFN antiviral response is known to be directly coupled to mevalonate-sterol biosynthesis, mechanistic insight for providing host pathway-therapeutic targets remain incomplete. Here, we show that Nampt and Sirt6 are coordinately regulated upon immune activation of macrophages and contribute to the IFN-sterol antiviral response. In silico analysis of the Nampt and Sirt6 promoter regions identified multiple core immune gene-regulatory transcription factor sites, including Stat1, implicating a molecular link to IFN control. Experimentally, we show using a range of genetically IFN-defective macrophages that the expression of Nampt is stringently regulated by the Jak/Stat-pathway while Sirt6 activation is temporally displaced in a partial IFN-dependent manner. We further show that pharmacological inhibition of Nampt and small interfering RNA (siRNA)-mediated inhibition of Nampt and Sirt6 promotes viral growth of cytomegalovirus in both fibroblasts and macrophages. Our results support the notion of pharmacologically exploiting immune regulated enzyme systems of macrophages for use as an adjuvant-based therapy for augmenting host protective pathway responses to infection.

2.
Nat Commun ; 10(1): 498, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700717

ABSTRACT

The mechanisms controlling CD4+ T cell switching from an effector to an anti-inflammatory (IL-10+) phenotype play an important role in the persistence of chronic inflammatory diseases. Here, we identify the cholesterol biosynthesis pathway as a key regulator of this process. Pathway analysis of cultured cytokine-producing human T cells reveals a significant association between IL-10 and cholesterol metabolism gene expression. Inhibition of the cholesterol biosynthesis pathway with atorvastatin or 25-hydroxycholesterol during switching from IFNγ+ to IL-10+ shows a specific block in immune resolution, defined as a significant decrease in IL-10 expression. Mechanistically, the master transcriptional regulator of IL10 in T cells, c-Maf, is significantly decreased by physiological levels of 25-hydroxycholesterol. Strikingly, progression to rheumatoid arthritis is associated with altered expression of cholesterol biosynthesis genes in synovial biopsies of predisposed individuals. Our data reveal a link between sterol metabolism and the regulation of the anti-inflammatory response in human CD4+ T cells.


Subject(s)
Cholesterol/biosynthesis , Interferon-gamma/metabolism , Interleukin-10/metabolism , Th1 Cells/metabolism , Atorvastatin/pharmacology , Humans , Hydroxycholesterols/pharmacology , Th1 Cells/drug effects
3.
PLoS Biol ; 14(3): e1002364, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26938778

ABSTRACT

In invertebrates, small interfering RNAs are at the vanguard of cell-autonomous antiviral immunity. In contrast, antiviral mechanisms initiated by interferon (IFN) signaling predominate in mammals. Whilst mammalian IFN-induced miRNA are known to inhibit specific viruses, it is not known whether host-directed microRNAs, downstream of IFN-signaling, have a role in mediating broad antiviral resistance. By performing an integrative, systematic, global analysis of RNA turnover utilizing 4-thiouridine labeling of newly transcribed RNA and pri/pre-miRNA in IFN-activated macrophages, we identify a new post-transcriptional viral defense mechanism mediated by miR-342-5p. On the basis of ChIP and site-directed promoter mutagenesis experiments, we find the synthesis of miR-342-5p is coupled to the antiviral IFN response via the IFN-induced transcription factor, IRF1. Strikingly, we find miR-342-5p targets mevalonate-sterol biosynthesis using a multihit mechanism suppressing the pathway at different functional levels: transcriptionally via SREBF2, post-transcriptionally via miR-33, and enzymatically via IDI1 and SC4MOL. Mass spectrometry-based lipidomics and enzymatic assays demonstrate the targeting mechanisms reduce intermediate sterol pathway metabolites and total cholesterol in macrophages. These results reveal a previously unrecognized mechanism by which IFN regulates the sterol pathway. The sterol pathway is known to be an integral part of the macrophage IFN antiviral response, and we show that miR-342-5p exerts broad antiviral effects against multiple, unrelated pathogenic viruses such Cytomegalovirus and Influenza A (H1N1). Metabolic rescue experiments confirm the specificity of these effects and demonstrate that unrelated viruses have differential mevalonate and sterol pathway requirements for their replication. This study, therefore, advances the general concept of broad antiviral defense through multihit targeting of a single host pathway.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferons/physiology , MicroRNAs/metabolism , Sterols/biosynthesis , Virus Diseases/immunology , Animals , Mice, Inbred C57BL
4.
Front Immunol ; 7: 634, 2016.
Article in English | MEDLINE | ID: mdl-28066443

ABSTRACT

The sterol metabolic network is emerging center stage in inflammation and immunity. Historically, observational clinical studies show that hypocholesterolemia is a common side effect of interferon (IFN) treatment. More recently, comprehensive systems-wide investigations of the macrophage IFN response reveal a direct molecular link between cholesterol metabolism and infection. Upon infection, flux through the sterol metabolic network is acutely moderated by the IFN response at multiple regulatory levels. The precise mechanisms by which IFN regulates the mevalonate-sterol pathway-the spine of the network-are beginning to be unraveled. In this review, we discuss our current understanding of the multifactorial mechanisms by which IFN regulates the sterol pathway. We also consider bidirectional communications resulting in sterol metabolism regulation of immunity. Finally, we deliberate on how this fundamental interaction functions as an integral element of host protective responses to infection and harmful inflammation.

5.
Steroids ; 99(Pt B): 219-29, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25759117

ABSTRACT

Interferons (IFNs) play a central role in immunity and emerging evidence suggests that IFN-signalling coordinately regulates sterol biosynthesis in macrophages, via Sterol Regulatory Element-Binding Protein (SREBP) dependent and independent pathways. However, the precise mechanisms and kinetic steps by which IFN controls sterol biosynthesis are as yet not fully understood. Here, we elucidate the molecular circuitry governing how IFN controls the first regulated step in the mevalonate-sterol pathway, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), through the synthesis of 25-Hydroxycholesterol (25-HC) from cholesterol by the IFN-inducible Cholesterol-25-Hydroxylase (CH25H). We show for the first 30-min of IFN stimulation of macrophages the rate of de novo synthesis of the Ch25h transcript is markedly increased but by 120-min becomes transcriptionally curtailed, coincident with induction of the Activating Transcription Factor 3 (ATF3) repressor. We demonstrate ATF3 induction by Toll-like receptors is strictly dependent on IFN-signalling. While the SREBP-pathway dependent rates of de novo transcription of Hmgcr are relatively unchanged in the first 90-min of IFN treatment, we find HMGCR enzyme levels undergo a rapid proteasomal-mediated degradation, defining a previously unappreciated SREBP-independent mechanism for IFN-action. These events precede a sustained marked reduction in Hmgcr RNA levels involving SREBP-dependent mechanisms. We demonstrate that HMGCR proteasomal-degradation by IFN strictly requires the synthesis of endogenous 25-HC and functionally couples HMGCR to CH25H to coordinately suppress sterol biosynthesis. In conclusion, we quantitatively delineate proteomic and transcriptional levels of IFN-mediated control of HMGCR, the primary enzymatic step of the mevalonate-sterol biosynthesis pathway, providing a foundational framework for mathematically modelling the therapeutic outcome of immune-metabolic pathways.


Subject(s)
Hydroxycholesterols/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Interferon-gamma/pharmacology , Macrophages/metabolism , Proteasome Endopeptidase Complex/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Kinetics , Macrophages/drug effects , Mice, Inbred C57BL , Models, Biological , Proteolysis/drug effects , Proteomics , RNA/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Time Factors , Transcription, Genetic/drug effects
6.
Immunity ; 38(1): 106-18, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23273843

ABSTRACT

Recent studies suggest that the sterol metabolic network participates in the interferon (IFN) antiviral response. However, the molecular mechanisms linking IFN with the sterol network and the identity of sterol mediators remain unknown. Here we report a cellular antiviral role for macrophage production of 25-hydroxycholesterol (cholest-5-en-3ß,25-diol, 25HC) as a component of the sterol metabolic network linked to the IFN response via Stat1. By utilizing quantitative metabolome profiling of all naturally occurring oxysterols upon infection or IFN-stimulation, we reveal 25HC as the only macrophage-synthesized and -secreted oxysterol. We show that 25HC can act at multiple levels as a potent paracrine inhibitor of viral infection for a broad range of viruses. We also demonstrate, using transcriptional regulatory-network analyses, genetic interventions and chromatin immunoprecipitation experiments that Stat1 directly coupled Ch25h regulation to IFN in macrophages. Our studies describe a physiological role for 25HC as a sterol-lipid effector of an innate immune pathway.


Subject(s)
Antiviral Agents/pharmacology , Hydroxycholesterols/metabolism , Interferons/pharmacology , Macrophages/immunology , Macrophages/metabolism , STAT1 Transcription Factor/metabolism , Animals , Binding Sites , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Gene Expression Regulation , Hydroxycholesterols/pharmacology , Liver X Receptors , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/virology , Mevalonic Acid/metabolism , Mice , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic , Protein Binding , Steroid Hydroxylases/genetics , Virus Replication/drug effects
7.
Biochimie ; 95(3): 613-21, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22664637

ABSTRACT

The cholesterol biosynthesis pathway has recently been shown to play an important role in the innate immune response to viral infection with host protection occurring through a coordinate down regulation of the enzymes catalysing each metabolic step. In contrast, statin based drugs, which form the principle pharmaceutical agents for decreasing the activity of this pathway, target a single enzyme. Here, we build an ordinary differential equation model of the cholesterol biosynthesis pathway in order to investigate how the two regulatory strategies impact upon the behaviour of the pathway. We employ a modest set of assumptions: that the pathway operates away from saturation, that each metabolite is involved in multiple cellular interactions and that mRNA levels reflect enzyme concentrations. Using data taken from primary bone marrow derived macrophage cells infected with murine cytomegalovirus or treated with IFNγ, we show that, under these assumptions, coordinate down-regulation of enzyme activity imparts a graduated reduction in flux along the pathway. In contrast, modelling a statin-like treatment that achieves the same degree of down-regulation in cholesterol production, we show that this delivers a step change in flux along the pathway. The graduated reduction mediated by physiological coordinate regulation of multiple enzymes supports a mechanism that allows a greater level of specificity, altering cholesterol levels with less impact upon interactions branching from the pathway, than pharmacological step reductions. We argue that coordinate regulation is likely to show a long-term evolutionary advantage over single enzyme regulation. Finally, the results from our models have implications for future pharmaceutical therapies intended to target cholesterol production with greater specificity and fewer off target effects, suggesting that this can be achieved by mimicking the coordinated down-regulation observed in immunological responses.


Subject(s)
Cholesterol/biosynthesis , Cholesterol/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity/drug effects , Models, Biological , Immunity, Innate/drug effects , Interferon-gamma/pharmacology , Muromegalovirus/physiology , Reproducibility of Results
8.
OMICS ; 16(5): 245-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22385281

ABSTRACT

Cells exploit signaling pathways during responses to environmental changes, and these processes are often modulated during disease. Particularly, relevant human pathologies such as cancer or viral infections require downregulating apoptosis signaling pathways to progress. As a result, the identification of proteins responsible for these changes is essential for the diagnostics and development of therapeutics. Transferring functional annotation within protein interaction networks has proven useful to identify such proteins, although this is not a trivial task. Here, we used different scoring methods to transfer annotation from 53 well-studied members of the human apoptosis pathways (as known by 2005) to their protein interactors. All scoring methods produced significant predictions (compared to a random negative model), but its number was too large to be useful. Thus, we made a final prediction using specific combinations of scoring methods and compared it to the proteins related to apoptosis signaling pathways during the last 5 years. We propose 273 candidate proteins that may be relevant in apoptosis signaling pathways. Although some of them have known functions consistent with their proposed apoptotsis involvement, the majority have not been annotated yet, leaving room for further experimental studies. We provide our predictions at http://sbi.imim.es/web/Apoptosis.php.


Subject(s)
Apoptosis/physiology , Protein Interaction Maps/physiology , Signal Transduction/physiology , Computational Biology/methods , Humans , Models, Biological
9.
J Virol ; 85(19): 10286-99, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775459

ABSTRACT

Activated macrophages play a central role in controlling inflammatory responses to infection and are tightly regulated to rapidly mount responses to infectious challenge. Type I interferon (alpha/beta interferon [IFN-α/ß]) and type II interferon (IFN-γ) play a crucial role in activating macrophages and subsequently restricting viral infections. Both types of IFNs signal through related but distinct signaling pathways, inducing a vast number of interferon-stimulated genes that are overlapping but distinguishable. The exact mechanism by which IFNs, particularly IFN-γ, inhibit DNA viruses such as cytomegalovirus (CMV) is still not fully understood. Here, we investigate the antiviral state developed in macrophages upon reversible inhibition of murine CMV by IFN-γ. On the basis of molecular profiling of the reversible inhibition, we identify a significant contribution of a restricted type I IFN subnetwork linked with IFN-γ activation. Genetic knockout of the type I-signaling pathway, in the context of IFN-γ stimulation, revealed an essential requirement for a primed type I-signaling process in developing a full refractory state in macrophages. A minimal transient induction of IFN-ß upon macrophage activation with IFN-γ is also detectable. In dose and kinetic viral replication inhibition experiments with IFN-γ, the establishment of an antiviral effect is demonstrated to occur within the first hours of infection. We show that the inhibitory mechanisms at these very early times involve a blockade of the viral major immediate-early promoter activity. Altogether our results show that a primed type I IFN subnetwork contributes to an immediate-early antiviral state induced by type II IFN activation of macrophages, with a potential further amplification loop contributed by transient induction of IFN-ß.


Subject(s)
Interferon Type I/immunology , Interferon-gamma/immunology , Macrophages/immunology , Macrophages/virology , Muromegalovirus/growth & development , Muromegalovirus/immunology , Animals , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , Time Factors
10.
PLoS Biol ; 9(3): e1000598, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21408089

ABSTRACT

Little is known about the protective role of inflammatory processes in modulating lipid metabolism in infection. Here we report an intimate link between the innate immune response to infection and regulation of the sterol metabolic network characterized by down-regulation of sterol biosynthesis by an interferon regulatory loop mechanism. In time-series experiments profiling genome-wide lipid-associated gene expression of macrophages, we show a selective and coordinated negative regulation of the complete sterol pathway upon viral infection or cytokine treatment with IFNγ or ß but not TNF, IL1ß, or IL6. Quantitative analysis at the protein level of selected sterol metabolic enzymes upon infection shows a similar level of suppression. Experimental testing of sterol metabolite levels using lipidomic-based measurements shows a reduction in metabolic output. On the basis of pharmacologic and RNAi inhibition of the sterol pathway we show augmented protection against viral infection, and in combination with metabolite rescue experiments, we identify the requirement of the mevalonate-isoprenoid branch of the sterol metabolic network in the protective response upon statin or IFNß treatment. Conditioned media experiments from infected cells support an involvement of secreted type 1 interferon(s) to be sufficient for reducing the sterol pathway upon infection. Moreover, we show that infection of primary macrophages containing a genetic knockout of the major type I interferon, IFNß, leads to only a partial suppression of the sterol pathway, while genetic knockout of the receptor for all type I interferon family members, ifnar1, or associated signaling component, tyk2, completely abolishes the reduction of the sterol biosynthetic activity upon infection. Levels of the proteolytically cleaved nuclear forms of SREBP2, a key transcriptional regulator of sterol biosynthesis, are reduced upon infection and IFNß treatment at both the protein and de novo transcription level. The reduction in srebf2 gene transcription upon infection and IFN treatment is also found to be strictly dependent on ifnar1. Altogether these results show that type 1 IFN signaling is both necessary and sufficient for reducing the sterol metabolic network activity upon infection, thereby linking the regulation of the sterol pathway with interferon anti-viral defense responses. These findings bring a new link between sterol metabolism and interferon antiviral response and support the idea of using host metabolic modifiers of innate immunity as a potential antiviral strategy.


Subject(s)
Down-Regulation , Herpesviridae Infections/immunology , Interferon-beta/physiology , Interferon-gamma/physiology , Muromegalovirus/immunology , Sterols/biosynthesis , Animals , Antiviral Agents/pharmacology , Cholesterol/metabolism , Herpesviridae Infections/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/pharmacology , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NIH 3T3 Cells , RNA Interference , Signal Transduction , Simvastatin/pharmacology , Sterol Regulatory Element Binding Protein 2/physiology
11.
BMC Syst Biol ; 2: 36, 2008 Apr 23.
Article in English | MEDLINE | ID: mdl-18433497

ABSTRACT

BACKGROUND: The complex yet flexible cellular response to pathogens is orchestrated by the interaction of multiple signalling and metabolic pathways. The molecular regulation of this response has been studied in great detail but comprehensive and unambiguous diagrams describing these events are generally unavailable. Four key signalling cascades triggered early-on in the innate immune response are the toll-like receptor, interferon, NF-kappaB and apoptotic pathways, which co-operate to defend cells against a given pathogen. However, these pathways are commonly viewed as separate entities rather than an integrated network of molecular interactions. RESULTS: Here we describe the construction of a logically represented pathway diagram which attempts to integrate these four pathways central to innate immunity using a modified version of the Edinburgh Pathway Notation. The pathway map is available in a number of electronic formats and editing is supported by yEd graph editor software. CONCLUSION: The map presents a powerful visual aid for interpreting the available pathway interaction knowledge and underscores the valuable contribution well constructed pathway diagrams make to communicating large amounts of molecular interaction data. Furthermore, we discuss issues with the limitations and scalability of pathways presented in this fashion, explore options for automated layout of large pathway networks and demonstrate how such maps can aid the interpretation of functional studies.


Subject(s)
Macrophage Activation/physiology , Models, Biological , Signal Transduction/immunology , Systems Biology/methods , Animals , Audiovisual Aids , Bone Marrow , Cells, Cultured , Computer Graphics , Immunity, Innate/physiology , Interferons/metabolism , Logic , Male , Mice , Mice, Inbred BALB C , Monocytes/immunology , NF-kappa B/metabolism , Protein Interaction Mapping/methods , Systems Integration , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Am Med Inform Assoc ; 15(2): 246-54, 2008.
Article in English | MEDLINE | ID: mdl-18096914

ABSTRACT

OBJECTIVE: The European INFOBIOMED Network of Excellence recognized that a successful education program in biomedical informatics should include not only traditional teaching activities in the basic sciences but also the development of skills for working in multidisciplinary teams. DESIGN: A carefully developed 3-year training program for biomedical informatics students addressed these educational aspects through the following four activities: (1) an internet course database containing an overview of all Medical Informatics and BioInformatics courses, (2) a BioMedical Informatics Summer School, (3) a mobility program based on a 'brokerage service' which published demands and offers, including funding for research exchange projects, and (4) training challenges aimed at the development of multi-disciplinary skills. MEASUREMENTS: This paper focuses on experiences gained in the development of novel educational activities addressing work in multidisciplinary teams. The training challenges described here were evaluated by asking participants to fill out forms with Likert scale based questions. For the mobility program a needs assessment was carried out. RESULTS: The mobility program supported 20 exchanges which fostered new BMI research, resulted in a number of peer-reviewed publications and demonstrated the feasibility of this multidisciplinary BMI approach within the European Union. Students unanimously indicated that the training challenge experience had contributed to their understanding and appreciation of multidisciplinary teamwork. CONCLUSION: The training activities undertaken in INFOBIOMED have contributed to a multi-disciplinary BMI approach. It is our hope that this work might provide an impetus for training efforts in Europe, and yield a new generation of biomedical informaticians.


Subject(s)
Medical Informatics/education , Consumer Behavior , Data Collection , European Union , Interdisciplinary Communication , Medical Informatics/economics , Program Evaluation
13.
J Virol ; 81(24): 13761-70, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928340

ABSTRACT

The prevalence and importance of microRNAs (miRNAs) in viral infection are increasingly relevant. Eleven miRNAs were previously identified in human cytomegalovirus (HCMV); however, miRNA content in murine CMV (MCMV), which serves as an important in vivo model for CMV infection, has not previously been examined. We have cloned and characterized 17 novel miRNAs that originate from at least 12 precursor miRNAs in MCMV and are not homologous to HCMV miRNAs. In parallel, we applied a computational analysis, using a support vector machine approach, to identify potential precursor miRNAs in MCMV. Four of the top 10 predicted precursor sequences were cloned in this study, and the combination of computational and cloning analysis demonstrates that MCMV has the capacity to encode miRNAs clustered throughout the genome. On the basis of drug sensitivity experiments for resolving the kinetic class of expression, we show that the MCMV miRNAs are both early and late gene products. Notably, the MCMV miRNAs occur on complementary strands of the genome in specific regions, a feature which has not previously been observed for viral miRNAs. One cluster of miRNAs occurs in close proximity to the 5' splice site of the previously identified 7.2-kb stable intron, implying a variety of potential regulatory mechanisms for MCMV miRNAs.


Subject(s)
Genome, Viral , Introns/genetics , Macrophages/virology , MicroRNAs/genetics , Muromegalovirus/genetics , RNA, Complementary/genetics , Animals , Base Sequence , Bone Marrow Cells , Cells, Cultured , Cloning, Molecular , Computational Biology/methods , Fibroblasts/virology , L Cells , Male , Mice , Mice, Inbred BALB C , MicroRNAs/analysis , MicroRNAs/isolation & purification , Molecular Sequence Data , NIH 3T3 Cells/virology , RNA, Viral/analysis , RNA, Viral/genetics , RNA, Viral/isolation & purification
14.
Mol Cell Proteomics ; 6(8): 1318-30, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17470424

ABSTRACT

Non-somatic synaptic and axonal compartments of neurons are primary pathological targets in many neurodegenerative conditions, ranging from Alzheimer disease through to motor neuron disease. Axons and synapses are protected from degeneration by the slow Wallerian degeneration (Wld(s)) gene. Significantly the molecular mechanisms through which this spontaneous genetic mutation delays degeneration remain controversial, and the downstream protein targets of Wld(s) resident in non-somatic compartments remain unknown. In this study we used differential proteomics analysis to identify proteins whose expression levels were significantly altered in isolated synaptic preparations from the striatum of Wld(s) mice. Eight of the 16 proteins we identified as having modified expression levels in Wld(s) synapses are known regulators of mitochondrial stability and degeneration (including VDAC1, Aralar1, and mitofilin). Subsequent analyses demonstrated that other key mitochondrial proteins, not identified in our initial screen, are also modified in Wld(s) synapses. Of the non-mitochondrial proteins identified, several have been implicated in neurodegenerative diseases where synapses and axons are primary pathological targets (including DRP-2 and Rab GDP dissociation inhibitor beta). In addition, we show that downstream protein changes can be identified in pathways corresponding to both Ube4b (including UBE1) and Nmnat1 (including VDAC1 and Aralar1) components of the chimeric Wld(s) gene, suggesting that full-length Wld(s) protein is required to elicit maximal changes in synaptic proteins. We conclude that altered mitochondrial responses to degenerative stimuli are likely to play an important role in the neuroprotective Wld(s) phenotype and that targeting proteins identified in the current study may lead to novel therapies for the treatment of neurodegenerative diseases in humans.


Subject(s)
Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Wallerian Degeneration/metabolism , Animals , Female , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Proteomics
15.
Am J Obstet Gynecol ; 195(2): 406.e1-16, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890550

ABSTRACT

OBJECTIVE: The purpose of this study was to test whether a quantitative high-throughput molecular screen can be used to probe human endometrium and initiate the development of molecular diagnostic tools with potential for identification of therapeutic targets in women with menstrual complaints. STUDY DESIGN: Endometrium was collected from 10 patients with complaint of heavy bleeding, classified into mid or late secretory phase of the menstrual cycle by histologic dating and serum progesterone concentration. Total RNA was extracted and gene activity assessed using high-density oligonucleotide arrays. RESULTS: Statistical testing identified 83 'signature' genes whose expression levels differentiated the mid and late secretory phases of the menstrual cycle. CONCLUSION: The results show that the endometrium, a complex heterogeneous tissue, is amenable to high-throughput molecular analyses and this work provides further support for the future application of molecular profiling to clinical diagnosis.


Subject(s)
Endometrium/physiology , Gene Expression Profiling , Luteal Phase/genetics , Menorrhagia/genetics , Oligonucleotide Array Sequence Analysis , Adult , Biopsy , Female , Humans , Insulin-Like Growth Factor I/genetics , Leiomyoma/genetics , Matrix Metalloproteinase 10 , Metalloendopeptidases/genetics , Pelvic Pain/genetics , Progesterone/blood , Receptors, Endothelin/genetics , Receptors, Thrombin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Uterine Neoplasms/genetics
16.
BMC Genomics ; 6: 178, 2005 Dec 12.
Article in English | MEDLINE | ID: mdl-16343346

ABSTRACT

BACKGROUND: Macrophages play an integral role in the host immune system, bridging innate and adaptive immunity. As such, they are finely attuned to extracellular and intracellular stimuli and respond by rapidly initiating multiple signalling cascades with diverse effector functions. The macrophage cell is therefore an experimentally and clinically amenable biological system for the mapping of biological pathways. The goal of the macrophage expression atlas is to systematically investigate the pathway biology and interaction network of macrophages challenged with a variety of insults, in particular via infection and activation with key inflammatory mediators. As an important first step towards this we present a single searchable database resource containing high-throughput macrophage gene expression studies. DESCRIPTION: The GPX Macrophage Expression Atlas (GPX-MEA) is an online resource for gene expression based studies of a range of macrophage cell types following treatment with pathogens and immune modulators. GPX-MEA follows the MIAME standard and includes an objective quality score with each experiment. It places special emphasis on rigorously capturing the experimental design and enables the searching of expression data from different microarray experiments. Studies may be queried on the basis of experimental parameters, sample information and quality assessment score. The ability to compare the expression values of individual genes across multiple experiments is provided. In addition, the database offers access to experimental annotation and analysis files and includes experiments and raw data previously unavailable to the research community. CONCLUSION: GPX-MEA is the first example of a quality scored gene expression database focussed on a macrophage cellular system that allows efficient identification of transcriptional patterns. The resource will provide novel insights into the phenotypic response of macrophages to a variety of benign, inflammatory, and pathogen insults. GPX-MEA is available through the GPX website at http://www.gti.ed.ac.uk/GPX.


Subject(s)
Computational Biology , Databases, Genetic , Gene Expression Profiling , Macrophages/chemistry , Animals , Data Collection , Humans , Microarray Analysis/methods , Quality Control , Research Design , Software Design
17.
Infect Immun ; 72(10): 6076-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385512

ABSTRACT

Infection with intestinal nematodes induces profound pathological changes to the gut that are associated with eventual parasite expulsion. We have applied expression profiling as an initial screening process with oligonucleotide microarrays (Affymetrix MG-U74AV2 gene chips) and time course kinetics to investigate gene transcription triggered by the intraepithelial nematode Trichinella spiralis in jejunal epithelium from BALB/c mice. Of the 4,114 genes detected, 2,617 were present in all uninfected and T. spiralis-infected replicates, 8% of which were notably upregulated, whereas 12% were downregulated at the time of worm expulsion (day 14 postinfection). Upregulation of goblet cell mucin gene transcripts intestinal mucin gene 3 (MUC3), calcium chloride channel 5 (CLCA5), and goblet cell gene 4 (GOB4) is consistent with enhanced production and alteration of mucus, whereas a 60- to 70-fold upregulation of transcripts for mast cell proteases 1 and 2 (MCPT-1 and -2) is consistent with intraepithelial mucosal mast cell recruitment. Importantly, there was novel expression of sialyltransferase 4C (SIAT4C), small proline-rich protein 2A (SPRR2A), and resistin-like molecule beta (RELMbeta) on day 14 postinfection. In contrast, DNase I and regenerating protein 3 (REG3) transcripts were substantially downregulated. Time course analyses revealed early (within 48 h of infection) induction of Siat4c, Sprr2A, and Relmbeta and later (within 120 h) induction of Mcpt-1 and -2. The findings demonstrate early innate responses and later inflammatory changes within the epithelium. The early epithelial responses may be associated both with repair (Sprr2A) and with the development of innate immunity (Siat4c and Relmbeta).


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/parasitology , Gene Expression Profiling , Inflammation/genetics , Jejunum/cytology , Trichinella spiralis/physiology , Trichinellosis/genetics , Animals , Antioxidants/metabolism , Cytoskeleton/genetics , Cytoskeleton/parasitology , Epithelial Cells/cytology , Epithelial Cells/enzymology , Female , Gene Expression Regulation , Glutathione/metabolism , Goblet Cells/metabolism , Goblet Cells/parasitology , Immunity/genetics , Inflammation/parasitology , Jejunum/enzymology , Jejunum/metabolism , Jejunum/parasitology , Male , Mast Cells/metabolism , Mast Cells/parasitology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mucins/biosynthesis , Oligonucleotide Array Sequence Analysis , Organ Specificity , Paneth Cells/metabolism , Paneth Cells/parasitology , Tight Junctions/genetics , Tight Junctions/parasitology , Transcription, Genetic/genetics , Trichinellosis/enzymology , Trichinellosis/metabolism
18.
Vaccine ; 22(11-12): 1433-40, 2004 Mar 29.
Article in English | MEDLINE | ID: mdl-15063566

ABSTRACT

Human gammaherpesviruses such as Epstein-Barr virus (EBV) cause lifelong infections and associated diseases, including malignancies, and the development of an effective vaccine against this class of viral infections is of considerable interest. The murine herpesvirus 68 (MHV-68) model provides a useful experimental setting to investigate the immune response to gammaherpesvirus infections and to evaluate the efficacy of vaccination strategies. In this study, we tested a heat-inactivated MHV-68 vaccine in immunocompetent mice as well as in B cell-deficient or type I IFN receptor knockout mice. Vaccination with heat-inactivated MHV-68 protected immunocompetent mice from the acute MHV-68 infection in the lung and strongly reduced the expansion of latently infected cells in the spleen and the development of splenomegaly. A similar inhibition of the acute viral replication in the lung was also observed in vaccinated B cell-deficient mice. Of note, the inactivated MHV-68 vaccine completely protected type I IFN receptor knockout mice from the infection with a lethal dose of MHV-68.


Subject(s)
Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae/immunology , Herpesvirus Vaccines/immunology , Interferon Type I/physiology , Receptors, Interferon/physiology , Virus Latency/immunology , Virus Replication/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cell Line , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutralization Tests , Receptors, Interferon/genetics , Vaccination , Vaccines, Inactivated/immunology
19.
J Biol Chem ; 278(33): 31444-55, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12791696

ABSTRACT

Chronic exposure to lower oxygen tension may increase cellular resistance to different types of acute metabolic stress. Here, we show that 24-h-long exposure to slightly decreased oxygen tension (partial pressure of oxygen (PO2) of 100 mm Hg instead of normal 144 mm Hg) confers resistance against acute hypoxia/reoxygenation-induced Ca2+ loading in heart-derived H9c2 cells. The number of ATP-sensitive K+ (K(ATP)) channels were increased in cells exposed to PO2 = 100 mm Hg relative to cells exposed to PO2 = 144 mm Hg. This was due to an increase in transcription of SUR2A, a K(ATP) channel regulatory subunit, but not Kir6.2, a K(ATP) channel pore-forming subunit. PO2 = 100 mm Hg also increased the SUR2 gene promoter activity. Experiments with cells overexpressing wild type of hypoxia-inducible factor (HIF)-1alpha and dominant negative HIF-1beta suggested that the HIF-1-signaling pathway did not participate in observed PO2-mediated regulation of SUR2A expression. On the other hand, NADH inhibited the effect of PO2 = 100 mm Hg but not the effect of PO2 = 20 mm Hg. LY 294002 and PD 184 352 prevented PO2-mediated regulation of K(ATP) channels, whereas rapamycin was without any effect. HMR 1098 inhibited the cytoprotective effect of PO2 = 100 mm Hg, and a decrease of PO2 from 144 to 100 mm Hg did not change the expression of any other gene, including those involved in stress and hypoxic response, as revealed by Affymetrix high density oligonucleotide arrays. We conclude that slight hypoxia activates HIF-1alpha-independent signaling cascade leading to an increase in SUR2A protein, a higher density of K(ATP) channels, and a cellular phenotype more resistant to acute metabolic stress.


Subject(s)
Hypoxia/metabolism , Myocardial Reperfusion Injury/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Acute Disease , Adenosine Triphosphate/metabolism , Animals , Calcium/pharmacology , Cell Membrane/metabolism , Cells, Cultured , Chronic Disease , Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocardium/cytology , NAD/metabolism , Oxygen/pharmacology , Phenotype , Promoter Regions, Genetic/physiology , Protein Serine-Threonine Kinases/metabolism , Rats , Sarcolemma/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...