51
|
A derived network-based interferon-related signature of human macrophages responding to Mycobacterium tuberculosis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:713071. [PMID: 25371902 PMCID: PMC4209755 DOI: 10.1155/2014/713071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 12/11/2022]
Abstract
Network analysis of transcriptional signature typically relies on direct interaction between two highly expressed genes. However, this approach misses indirect and biological relevant interactions through a third factor (hub). Here we determine whether a hub-based network analysis can select an improved signature subset that correlates with a biological change in a stronger manner than the original signature. We have previously reported an interferon-related transcriptional signature (THP1r2Mtb-induced) from Mycobacterium tuberculosis (M. tb)-infected THP-1 human macrophage. We selected hub-connected THP1r2Mtb-induced genes into the refined network signature TMtb-iNet and grouped the excluded genes into the excluded signature TMtb-iEx. TMtb-iNet retained the enrichment of binding sites of interferon-related transcription factors and contained relatively more interferon-related interacting genes when compared to THP1r2Mtb-induced signature. TMtb-iNet correlated as strongly as THP1r2Mtb-induced signature on a public transcriptional dataset of patients with pulmonary tuberculosis (PTB). TMtb-iNet correlated more strongly in CD4(+) and CD8(+) T cells from PTB patients than THP1r2Mtb-induced signature and TMtb-iEx. When TMtb-iNet was applied to data during clinical therapy of tuberculosis, it resulted in the most pronounced response and the weakest correlation. Correlation on dataset from patients with AIDS or malaria was stronger for TMtb-iNet, indicating an involvement of TMtb-iNet in these chronic human infections. Collectively, the significance of this work is twofold: (1) we disseminate a hub-based approach in generating a biologically meaningful and clinically useful signature; (2) using this approach we introduce a new network-based signature and demonstrate its promising applications in understanding host responses to infections.
Collapse
|
52
|
Zaas AK, Garner BH, Tsalik EL, Burke T, Woods CW, Ginsburg GS. The current epidemiology and clinical decisions surrounding acute respiratory infections. Trends Mol Med 2014; 20:579-88. [PMID: 25201713 DOI: 10.1016/j.molmed.2014.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 12/15/2022]
Abstract
Acute respiratory infection (ARI) is a common diagnosis in outpatient and emergent care settings. Currently available diagnostics are limited, creating uncertainty in the use of antibacterial, antiviral, or supportive care. Up to 72% of ambulatory care patients with ARI are treated with an antibacterial, despite only a small fraction actually needing one. Antibiotic overuse is not restricted to ambulatory care: ARI accounts for approximately 5 million emergency department (ED) visits annually in the USA, where 52-61% of such patients receive antibiotics. Thus, an accurate test for the presence or absence of viral or bacterial infection is needed. In this review, we focus on recent research showing that the host-response (genomic, proteomic, or miRNA) can accomplish this task.
Collapse
Affiliation(s)
- Aimee K Zaas
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| | - Bronwen H Garner
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Ephraim L Tsalik
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Thomas Burke
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Christopher W Woods
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
53
|
Xing D, Markey K, Das RG, Feavers I. Whole-cell pertussis vaccine potency assays: the Kendrick test and alternative assays. Expert Rev Vaccines 2014; 13:1175-82. [PMID: 25182836 DOI: 10.1586/14760584.2014.939636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Whole-cell pertussis vaccines are still widely used across the globe and have been shown to produce longer lasting immunity against pertussis infection than acellular pertussis vaccines. Therefore, whole-cell vaccines are likely to continue to be used for the foreseeable future. The intracerebral mouse protection test (Kendrick test) is effective for determining the potency of whole-cell pertussis vaccines and is the only test that has shown a correlation with protection in children. Here we review the Kendrick test in terms of international requirements for vaccine potency and critical technical points to be considered for a successful test including test validity, in-house references and statistical analysis. There are objections to the Kendrick test on animal welfare and technical grounds. Respiratory challenge assays, nitric oxide induction assay and serological assays have been developed and have been proposed as possible methods which might provide alternatives to the Kendrick test. These methods and their limitations are also briefly discussed. Establishment of validated in vitro correlates of protection has yet to be achieved. New technical developments, such as genome sequence and the use of gene microarrays to screen responses triggered by vaccine components may also provide leads to alternative assays to the Kendrick test by identifying biomarkers of protection.
Collapse
Affiliation(s)
- Dorothy Xing
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, EN6 3QG, UK
| | | | | | | |
Collapse
|
54
|
Abstract
ABSTRACT: The importance of macrophages in the control of infections has long been documented, but macrophages have also been shown to contribute to severe influenza A virus infections. Macrophage function ranges from highly proinflammatory to wound healing and regulatory and a picture of diverse subsets with considerable plasticity in function and phenotype is emerging. Within the lung three subsets of macrophage populations have been identified: resident alveolar macrophages, interstitial macrophages and exudate-derived macrophages. Here we review model systems and techniques for defining macrophage function in vivo and discuss macrophage infection in vitro. The use of detailed phenotypic approaches and techniques to dissect the role of individual macrophage subsets in vivo promises rapid advances in this area of research.
Collapse
Affiliation(s)
- Marlynne Q Nicol
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, UK
| | - Bernadette M Dutia
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, UK
| |
Collapse
|
55
|
Raza S, Barnett MW, Barnett-Itzhaki Z, Amit I, Hume DA, Freeman TC. Analysis of the transcriptional networks underpinning the activation of murine macrophages by inflammatory mediators. J Leukoc Biol 2014; 96:167-83. [PMID: 24721704 PMCID: PMC4378362 DOI: 10.1189/jlb.6hi0313-169r] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 01/17/2014] [Accepted: 02/23/2014] [Indexed: 01/09/2023] Open
Abstract
Macrophages respond to the TLR4 agonist LPS with a sequential transcriptional cascade controlled by a complex regulatory network of signaling pathways and transcription factors. At least two distinct pathways are currently known to be engaged by TLR4 and are distinguished by their dependence on the adaptor molecule MyD88. We have used gene expression microarrays to define the effects of each of three variables--LPS dose, LPS versus IFN-β and -γ, and genetic background--on the transcriptional response of mouse BMDMs. Analysis of correlation networks generated from the data has identified subnetworks or modules within the macrophage transcriptional network that are activated selectively by these variables. We have identified mouse strain-specific signatures, including a module enriched for SLE susceptibility candidates. In the modules of genes unique to different treatments, we found a module of genes induced by type-I IFN but not by LPS treatment, suggesting another layer of complexity in the LPS-TLR4 signaling feedback control. We also observe that the activation of the complement system, in common with the known activation of MHC class 2 genes, is reliant on IFN-γ signaling. Taken together, these data further highlight the exquisite nature of the regulatory systems that control macrophage activation, their likely relevance to disease resistance/susceptibility, and the appropriate response of these cells to proinflammatory stimuli.
Collapse
Affiliation(s)
- Sobia Raza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | - Mark W Barnett
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | | | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | - Tom C Freeman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| |
Collapse
|
56
|
Du Z, Yang H, Tan Y, Tian G, Zhang Q, Cui Y, Yanfeng Yan, Wu X, Chen Z, Cao S, Bi Y, Han Y, Wang X, Song Y, Yang R. Transcriptomic response to Yersinia pestis: RIG-I like receptor signaling response is detrimental to the host against plague. J Genet Genomics 2014; 41:379-96. [PMID: 25064677 DOI: 10.1016/j.jgg.2014.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/09/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023]
Abstract
Bacterial pathogens have evolved various mechanisms to modulate host immune responses for successful infection. In this study, RNA-sequencing technology was used to analyze the responses of human monocytes THP1 to Yersinia pestis infection. Over 6000 genes were differentially expressed over the 12 h infection. Kinetic responses of pathogen recognition receptor signaling pathways, apoptosis, antigen processing, and presentation pathway and coagulation system were analyzed in detail. Among them, RIG-I-like receptor (RLR) signaling pathway, which was established for antiviral defense, was significantly affected. Mice lacking MAVS, the adaptor of the RLR signaling pathway, were less sensitive to infection and exhibited lower IFN-β production, higher Th1-type cytokines IFN-γ and IL-12 production, and lower Th2-type cytokines IL-4 and IL-13 production in the serum compared with wild-type mice. Moreover, infection of pathogenic bacteria other than Y. pestis also altered the expression of the RLR pathway, suggesting that the response of RLR pathway to bacterial infection is a universal mechanism.
Collapse
Affiliation(s)
- Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Huiying Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yafang Tan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Guang Tian
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qingwen Zhang
- Qinghai Institute for Endemic Disease Prevention and Control of Qinghai Province, Xining 811602, China
| | - Yujun Cui
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yanfeng Yan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaohong Wu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | - Shiyang Cao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yanping Han
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaoyi Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing 100071, China.
| |
Collapse
|
57
|
Gorvel L, Textoris J, Banchereau R, Ben Amara A, Tantibhedhyangkul W, von Bargen K, Ka MB, Capo C, Ghigo E, Gorvel JP, Mege JL. Intracellular bacteria interfere with dendritic cell functions: role of the type I interferon pathway. PLoS One 2014; 9:e99420. [PMID: 24915541 PMCID: PMC4051653 DOI: 10.1371/journal.pone.0099420] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/14/2014] [Indexed: 01/18/2023] Open
Abstract
Dendritic cells (DCs) orchestrate host defenses against microorganisms. In infectious diseases due to intracellular bacteria, the inefficiency of the immune system to eradicate microorganisms has been attributed to the hijacking of DC functions. In this study, we selected intracellular bacterial pathogens with distinct lifestyles and explored the responses of monocyte-derived DCs (moDCs). Using lipopolysaccharide as a control, we found that Orientia tsutsugamushi, the causative agent of scrub typhus that survives in the cytosol of target cells, induced moDC maturation, as assessed by decreased endocytosis activity, the ability to induce lymphocyte proliferation and the membrane expression of phenotypic markers. In contrast, Coxiella burnetii, the agent of Q fever, and Brucella abortus, the agent of brucellosis, both of which reside in vacuolar compartments, only partly induced the maturation of moDCs, as demonstrated by a phenotypic analysis. To analyze the mechanisms used by C. burnetii and B. abortus to alter moDC activation, we performed microarray and found that C. burnetii and B. abortus induced a specific signature consisting of TLR4, TLR3, STAT1 and interferon response genes. These genes were down-modulated in response to C. burnetii and B. abortus but up-modulated in moDCs activated by lipopolysaccharide and O. tsutsugamushi. This transcriptional alteration was associated with the defective interferon-β production. This study demonstrates that intracellular bacteria specifically affect moDC responses and emphasizes how C. burnetii and B. abortus interfere with moDC activation and the antimicrobial immune response. We believe that comparing infection by several bacterial species may be useful for defining new pathways and biomarkers and for developing new treatment strategies.
Collapse
Affiliation(s)
- Laurent Gorvel
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Julien Textoris
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Romain Banchereau
- Baylor Institute for Immunology Research, Dallas, Texas, United States of America
| | - Amira Ben Amara
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Wiwit Tantibhedhyangkul
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
- Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kristin von Bargen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, INSERM, U1104, CNRS, UMR7280, Marseille, France
| | - Mignane B. Ka
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Christian Capo
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Eric Ghigo
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, INSERM, U1104, CNRS, UMR7280, Marseille, France
| | - Jean-Louis Mege
- Centre National de la Recherche Scientifique UMR 7278, IRD198, INSERM U1095, Aix-Marseille Université, Marseille, France
- * E-mail:
| |
Collapse
|
58
|
Samuels BA, Leonardo ED, Dranovsky A, Williams A, Wong E, Nesbitt AMI, McCurdy RD, Hen R, Alter M. Global state measures of the dentate gyrus gene expression system predict antidepressant-sensitive behaviors. PLoS One 2014; 9:e85136. [PMID: 24465494 PMCID: PMC3894967 DOI: 10.1371/journal.pone.0085136] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/23/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine are the most common form of medication treatment for major depression. However, approximately 50% of depressed patients fail to achieve an effective treatment response. Understanding how gene expression systems respond to treatments may be critical for understanding antidepressant resistance. METHODS We take a novel approach to this problem by demonstrating that the gene expression system of the dentate gyrus responds to fluoxetine (FLX), a commonly used antidepressant medication, in a stereotyped-manner involving changes in the expression levels of thousands of genes. The aggregate behavior of this large-scale systemic response was quantified with principal components analysis (PCA) yielding a single quantitative measure of the global gene expression system state. RESULTS Quantitative measures of system state were highly correlated with variability in levels of antidepressant-sensitive behaviors in a mouse model of depression treated with fluoxetine. Analysis of dorsal and ventral dentate samples in the same mice indicated that system state co-varied across these regions despite their reported functional differences. Aggregate measures of gene expression system state were very robust and remained unchanged when different microarray data processing algorithms were used and even when completely different sets of gene expression levels were used for their calculation. CONCLUSIONS System state measures provide a robust method to quantify and relate global gene expression system state variability to behavior and treatment. State variability also suggests that the diversity of reported changes in gene expression levels in response to treatments such as fluoxetine may represent different perspectives on unified but noisy global gene expression system state level responses. Studying regulation of gene expression systems at the state level may be useful in guiding new approaches to augmentation of traditional antidepressant treatments.
Collapse
Affiliation(s)
- Benjamin A. Samuels
- Departments of Psychiatry and Neuroscience, Columbia University, New York, New York, United States of America
| | - E. David Leonardo
- Departments of Psychiatry and Neuroscience, Columbia University, New York, New York, United States of America
| | - Alex Dranovsky
- Departments of Psychiatry and Neuroscience, Columbia University, New York, New York, United States of America
| | - Amanda Williams
- AstraZeneca Pharmaceuticals, CNS Discovery, Wilmington, Delaware, United States of America
| | - Erik Wong
- AstraZeneca Pharmaceuticals, CNS Discovery, Wilmington, Delaware, United States of America
| | - Addie May I. Nesbitt
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Richard D. McCurdy
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, New York, United States of America
- * E-mail: (MA); (RH)
| | - Mark Alter
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MA); (RH)
| |
Collapse
|
59
|
Abstract
Current methods for determining the potency and toxicity of pertussis vaccines are outdated and require improvement. The intracerebral challenge test is effective for determining the potency of whole-cell vaccines but is objectionable on animal welfare and technical grounds. The same applies to its modification for assaying acellular pertussis vaccines. Respiratory challenge methods offer an interim solution pending establishment of validated in vitro correlates of protection, for example nitric oxide induction. Their evaluation is being promoted by the World Health Organization through the Pertussis Vaccines Working Group. Current toxicity assays based on weight gain and histamine sensitization of mice are imprecise and need replacement. Limits need to be established for specific toxin content of both acellular and whole-cell vaccines and should be supported by specific assays. More precise methods based on determination of ribosyltransferase activity in tandem with receptor-binding assays are under evaluation. Genome sequence data and the use of gene microarrays to screen responses triggered by vaccine components may also provide leads to improved methods for assessing both toxicity and immunogenicity.
Collapse
Affiliation(s)
- Michael J Corbel
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, EN6 3QG, UK.
| | | |
Collapse
|
60
|
Frande-Cabanes E, Fernandez-Prieto L, Calderon-Gonzalez R, Rodríguez-Del Río E, Yañez-Diaz S, López-Fanarraga M, Alvarez-Domínguez C. Dissociation of innate immune responses in microglia infected with Listeria monocytogenes. Glia 2013; 62:233-46. [PMID: 24311463 PMCID: PMC4068285 DOI: 10.1002/glia.22602] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 10/30/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022]
Abstract
Microglia, the innate immune cells of the brain, plays a central role in cerebral listeriosis. Here, we present evidence that microglia control Listeria infection differently than macrophages. Infection of primary microglial cultures and murine cell lines with Listeria resulted in a dual function of the two gene expression programmes involved in early and late immune responses in macrophages. Whereas the bacterial gene hly seems responsible for both transcriptional programmes in macrophages, Listeria induces in microglia only the tumor necrosis factor (TNF)-regulated transcriptional programme. Listeria also represses in microglia the late immune response gathered in two clusters, microbial degradation, and interferon (IFN)-inducible genes. The bacterial gene actA was required in microglia to induce TNF-regulated responses and to repress the late response. Isolation of microglial phagosomes revealed a phagosomal environment unable to destroy Listeria. Microglial phagosomes were also defective in several signaling and trafficking components reported as relevant for Listeria innate immune responses. This transcriptional strategy in microglia induced high levels of TNF-α and monocyte chemotactic protein-1 and low production of other neurotoxic compounds such as nitric oxide, hydrogen peroxide, and Type I IFNs. These cytokines and toxic microglial products are also released by primary microglia, and this cytokine and chemokine cocktail display a low potential to trigger neuronal apoptosis. This overall bacterial strategy strongly suggests that microglia limit Listeria inflammation pattern exclusively through TNF-mediated responses to preserve brain integrity.
Collapse
Affiliation(s)
- Elisabet Frande-Cabanes
- Grupo de Genómica, Proteómica y vacunas, Instituto de Investigación y Formación Marqués de Valdecilla (IFIMAV), Primera Planta-Laboratorio 124, Avda. de Cardenal Herrera Oria, s/n, 39011, Santander, Spain
| | | | | | | | | | | | | |
Collapse
|
61
|
Crispo M, Van Maele L, Tabareau J, Cayet D, Errea A, Ferreira AM, Rumbo M, Sirard JC. Transgenic mouse model harboring the transcriptional fusion ccl20-luciferase as a novel reporter of pro-inflammatory response. PLoS One 2013; 8:e78447. [PMID: 24265691 PMCID: PMC3827052 DOI: 10.1371/journal.pone.0078447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/10/2013] [Indexed: 12/12/2022] Open
Abstract
The chemokine CCL20, the unique ligand of CCR6 functions as an attractant of immune cells. Expression of CCL20 is induced by Toll-like Receptor (TLR) signaling or proinflammatory cytokine stimulation. However CCL20 is also constitutively produced at specific epithelial sites of mucosa. This expression profile is achieved by transcriptional regulation. In the present work we characterized regulatory features of mouse Ccl20 gene. Transcriptional fusions between the mouse Ccl20 promoter and the firefly luciferase (luc) encoding gene were constructed and assessed in in vitro and in vivo assays. We found that liver CCL20 expression and luciferase activity were upregulated by systemic administration of the TLR5 agonist flagellin. Using shRNA and dominant negative form specific for mouse TLR5, we showed that this expression was controlled by TLR5. To address in situ the regulation of gene activity, a transgenic mouse line harboring a functional Ccl20-luc fusion was generated. The luciferase expression was highly concordant with Ccl20 expression in different tissues. Our data indicate that the transgenic mouse model can be used to monitor activation of innate response in vivo.
Collapse
Affiliation(s)
- Martina Crispo
- Unidad de Animales Transgénicos y de Experimentación – Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Laurye Van Maele
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Julien Tabareau
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Delphine Cayet
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
| | - Agustina Errea
- Laboratorio de Investigaciones del Sistema Inmune (LISIN) – National University of La Plata, Provincia de Buenos Aires, Argentina
| | - Ana María Ferreira
- Catedra de Inmunologia, Facultad de Ciencias/Facultad de Quimica, Universidad de la República, Montevideo, Uruguay
| | - Martin Rumbo
- Laboratorio de Investigaciones del Sistema Inmune (LISIN) – National University of La Plata, Provincia de Buenos Aires, Argentina
| | - Jean Claude Sirard
- Institut Pasteur de Lille, Centre d’Infection et d’Immunité de Lille, F-59000 Lille, Region Nord-Pas de Calais, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59000 Lille, Region Nord-Pas de Calais France
- Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, Region Nord-Pas de Calais France
- Univ Lille Nord de France, F-59000 Lille, Region Nord-Pas de Calais, France
- * E-mail:
| |
Collapse
|
62
|
Mejias A, Ramilo O. Transcriptional profiling in infectious diseases: ready for prime time? J Infect 2013; 68 Suppl 1:S94-9. [PMID: 24139187 DOI: 10.1016/j.jinf.2013.09.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2013] [Indexed: 11/19/2022]
Abstract
Blood represents a reservoir and a migration compartment of cells of the immune system. Traditional microbiologic diagnostic tests relied on laboratory identification of the pathogen causing the infection. However, this approach is less than optimal for a variety of reasons: pathogen's slow growth, resistance to cultivation in vitro or insufficient proof to establish causality when a pathogen is identified. An alternative approach to the pathogen-detection strategy is based on a comprehensive analysis of the host response to the infection by analysis of blood leukocytes gene expression profiles. This strategy has been successfully applied to distinguish and classify children and adults with acute infections caused by different pathogens. Molecular distance to health (MDTH) is a genomic score that measures the global transcriptional perturbation in each individual patient compared to healthy controls. Studies indicate that MDTH is a promising biomarker to help classifying patients according to clinical severity.
Collapse
Affiliation(s)
- Asuncion Mejias
- Division of Pediatric Infectious Diseases and Center for Vaccines and Immunity, Nationwide Children's Hospital and The Ohio State University, Columbus, OH 43205, USA
| | | |
Collapse
|
63
|
Xue F, Zhao X, Yang Y, Zhao J, Yang Y, Cao Y, Hong C, Liu Y, Sun L, Huang M, Gu J. Responses of murine and human macrophages to leptospiral infection: a study using comparative array analysis. PLoS Negl Trop Dis 2013; 7:e2477. [PMID: 24130911 PMCID: PMC3794915 DOI: 10.1371/journal.pntd.0002477] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 08/30/2013] [Indexed: 12/04/2022] Open
Abstract
Leptospirosis is a re-emerging tropical infectious disease caused by pathogenic Leptospira spp. The different host innate immune responses are partially related to the different severities of leptospirosis. In this study, we employed transcriptomics and cytokine arrays to comparatively calculate the responses of murine peritoneal macrophages (MPMs) and human peripheral blood monocytes (HBMs) to leptospiral infection. We uncovered a series of different expression profiles of these two immune cells. The percentages of regulated genes in several biological processes of MPMs, such as antigen processing and presentation, membrane potential regulation, and the innate immune response, etc., were much greater than those of HBMs (>2-fold). In MPMs and HBMs, the caspase-8 and Fas-associated protein with death domain (FADD)-like apoptosis regulator genes were significantly up-regulated, which supported previous results that the caspase-8 and caspase-3 pathways play an important role in macrophage apoptosis during leptospiral infection. In addition, the key component of the complement pathway, C3, was only up-regulated in MPMs. Furthermore, several cytokines, e.g. interleukin 10 (IL-10) and tumor necrosis factor alpha (TNF-alpha), were differentially expressed at both mRNA and protein levels in MPMs and HBMs. Some of the differential expressions were proved to be pathogenic Leptospira-specific regulations at mRNA level or protein level. Though it is still unclear why some animals are resistant and others are susceptible to leptospiral infection, this comparative study based on transcriptomics and cytokine arrays partially uncovered the differences of murine resistance and human susceptibility to leptospirosis. Taken together, these findings will facilitate further molecular studies on the innate immune response to leptospiral infection. Although pathogenic Leptospira is not an obligate intracellular pathogen, recent studies have shown that phagocytosis and innate immunity play important roles in leptospirosis. The Leptospira-macrophage interaction is a common model used to elucidate the initial response in leptospiral infection. Our previous research has shown that there is little difference in the transcriptomics of pathogenic Leptospira infecting murine or human macrophage cell lines. Contrarily, in this study, we observed significant differences of murine and human primary macrophages infected by L. interrogans as shown in several processes, such as antigen processing and presentation, Toll-like receptor signaling pathway and innate immune response, complement and coagulation cascades, expression of major cytokines and chemokines, etc. These results suggested that different immune responses explain the major disparities in the murine and human Leptospira-macrophage infection models. This study added to the former leptospiral transcriptomics research on the Leptospira-macrophage interaction model and laid a foundation for further investigation in the pathogenesis of leptospirosis.
Collapse
Affiliation(s)
- Feng Xue
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Tropical Medicine Research Institute, Beijing, China
- Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
- Beijing Institute of Biotechnology, Beijing, China
- * E-mail:
| | - Xinghui Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Yingchao Yang
- Division of Parasitic Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
| | - Jinping Zhao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yutao Yang
- Department of Neurobiology, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Major Brain Disorders, Beijing Institute of Brain Disorders, Beijing, China
| | - Yongguo Cao
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Cailing Hong
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yuan Liu
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lan Sun
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Tropical Medicine Research Institute, Beijing, China
- Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Minjun Huang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Tropical Medicine Research Institute, Beijing, China
- Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Junchao Gu
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Tropical Medicine Research Institute, Beijing, China
- Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| |
Collapse
|
64
|
Ma H, Zhong W, Jiang Y, Fontaine C, Li S, Fu J, Olkkonen VM, Staels B, Yan D. Increased atherosclerotic lesions in LDL receptor deficient mice with hematopoietic nuclear receptor Rev-erbα knock- down. J Am Heart Assoc 2013; 2:e000235. [PMID: 23963755 PMCID: PMC3828791 DOI: 10.1161/jaha.113.000235] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Nuclear receptor Rev-erbα plays important roles in circadian clock timing, lipid metabolism, adipogenesis, and vascular inflammation. However, the role of Rev-erbα in atherosclerotic lesion development has not been assessed in vivo. METHODS AND RESULTS The nuclear receptor Rev-erbα was knocked down in mouse haematopoietic cells by means of shRNA-lentiviral transduction, followed by bone marrow transplantation into LDL receptor knockout mice. The Rev-erbα protein in peripheral macrophage was reduced by 70% as compared to control mice injected with nontargeting shRNA lentivirus-transduced bone marrow. A significant increase in atherosclerotic lesions was observed around the aorta valves as well as upon en face aorta analysis of Rev-erbα knock-down bone marrow recipients (P<0.01) as compared to the control mice, while plasma cholesterol, phospholipid, and triacylglycerol levels were not affected. Overexpression of Rev-erbα in bone marrow mononuclear cells decreased inflammatory M1 while increasing M2 macrophage markers, while Rev-erbα knock down increased the macrophage inflammatory phenotype in vitro and in vivo. Furthermore, treatment of differentiating macrophages with the Rev-erbα ligand heme promoted expression of antiinflammatory M2 markers. CONCLUSIONS These observations identify hematopoietic cell Rev-erbα as a new modulator of atherogenesis in mice.
Collapse
Affiliation(s)
- Hongling Ma
- Department of Biotechnology, Jinan University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Eisenreich W, Heesemann J, Rudel T, Goebel W. Metabolic host responses to infection by intracellular bacterial pathogens. Front Cell Infect Microbiol 2013; 3:24. [PMID: 23847769 PMCID: PMC3705551 DOI: 10.3389/fcimb.2013.00024] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
The interaction of bacterial pathogens with mammalian hosts leads to a variety of physiological responses of the interacting partners aimed at an adaptation to the new situation. These responses include multiple metabolic changes in the affected host cells which are most obvious when the pathogen replicates within host cells as in case of intracellular bacterial pathogens. While the pathogen tries to deprive nutrients from the host cell, the host cell in return takes various metabolic countermeasures against the nutrient theft. During this conflicting interaction, the pathogen triggers metabolic host cell responses by means of common cell envelope components and specific virulence-associated factors. These host reactions generally promote replication of the pathogen. There is growing evidence that pathogen-specific factors may interfere in different ways with the complex regulatory network that controls the carbon and nitrogen metabolism of mammalian cells. The host cell defense answers include general metabolic reactions, like the generation of oxygen- and/or nitrogen-reactive species, and more specific measures aimed to prevent access to essential nutrients for the respective pathogen. Accurate results on metabolic host cell responses are often hampered by the use of cancer cell lines that already exhibit various de-regulated reactions in the primary carbon metabolism. Hence, there is an urgent need for cellular models that more closely reflect the in vivo infection conditions. The exact knowledge of the metabolic host cell responses may provide new interesting concepts for antibacterial therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Lehrstuhl für Biochemie, Center of Isotopologue Profiling, Technische Universität München Garching, Germany
| | | | | | | |
Collapse
|
66
|
Lindig S, Quickert S, Vodovotz Y, Wanner GA, Bauer M. Age-independent co-expression of antimicrobial gene clusters in the blood of septic patients. Int J Antimicrob Agents 2013; 42 Suppl:S2-7. [PMID: 23684387 DOI: 10.1016/j.ijantimicag.2013.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Recent research has unravelled the clinical potential of profiling the blood transcriptome to diagnose diseases. However, resulting molecular marker sets comprised features with varying robustness and performance, depending on the dimension of training data. Thus, we investigated patterns that are inherent in large-scale data and suitable for feature selection in application to blood samples from septic patients. By integrating >300 microarray samples in correlation and enrichment analysis, we found general response patterns including a vast majority of co-expressed genes. Differentially expressed genes significantly mapped to immune response-associated categories and revealed strongly correlating upregulated genes related to antimicrobial functions. Classifiers using >20 uncorrelated features from enriched functional categories performed with 85% correct classification on average (10-fold cross-validation), comparable with correlated features, whilst single genes achieved up to 83% correct classifications in identifying septic patients. Independent interplatform comparison, however, validated only a subset of these features, including the antimicrobial cluster (area under the receiver operating characteristic curve >0.8). Based on these results, we propose feature selection for classification incorporating correlation and enriched functional categories to obtain robust marker candidates. Results of this transcriptomic meta-analysis suggest age-independent diagnostic opportunities, although further observational and animal interventional experiments are required to confirm the relevance of antimicrobial genes in sepsis.
Collapse
Affiliation(s)
- Sandro Lindig
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Erlanger Allee 101, 07747 Jena, Germany
| | | | | | | | | |
Collapse
|
67
|
Gandoura S, Weiss E, Rautou PE, Fasseu M, Gustot T, Lemoine F, Hurtado-Nedelec M, Hego C, Vadrot N, Elkrief L, Lettéron P, Tellier Z, Pocidalo MA, Valla D, Lebrec D, Groyer A, Monteiro RC, de la Grange P, Moreau R. Gene- and exon-expression profiling reveals an extensive LPS-induced response in immune cells in patients with cirrhosis. J Hepatol 2013; 58:936-48. [PMID: 23321315 DOI: 10.1016/j.jhep.2012.12.025] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 12/26/2012] [Accepted: 12/28/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Lipopolysaccharide (LPS)-expressing bacteria cause severe inflammation in cirrhotic patients. The global gene response to LPS is unknown in cirrhotic immune cells. METHODS Gene-expression profiling using Affymetrix Human Exon Array analyzed the expression of 14,851 genes in LPS-stimulated peripheral blood mononuclear cells (PBMCs) from 4 patients with cirrhosis and 4 healthy subjects. We performed validation studies using RT-qPCR in LPS-stimulated PBMCs from 52 patients and 9 healthy subjects and investigated the association of gene induction with mortality in 26 patients. RESULTS Gene-expression profiling of LPS-stimulated cirrhotic cells showed 509 upregulated genes and 1588 downregulated genes. In LPS-stimulated "healthy" cells, 952 genes were upregulated and 838 genes downregulated. The 741 LPS-regulated genes shared by cirrhotic and "healthy" cells were involved in cytokine production/activity and induction of "immune paralysis". Comparison of functions associated with the 1356 genes, specifically regulated by LPS in cirrhotic cells, to functions of the 1049 genes, specifically regulated in "healthy" cells, allowed to define a cirrhosis-specific phenotype. Unlike in "healthy" cells, LPS failed to induce an interferon-mediated program in cirrhotic cells. In cirrhotic PBMCs, LPS specifically induced certain molecules involved in apoptosis and downregulated molecules involved in endocytic trafficking. RT-qPCR experiments showed that LPS-stimulated cirrhotic PBMCs had an enhanced induction of certain proinflammatory cytokines and chemokines. In the prognosis study, higher ex vivo LPS-induction of the inflammatory genes IL6 and CXCL5 was a significant predictor of mortality. CONCLUSIONS Our results show that LPS-stimulated cirrhotic PBMCs exhibit an extensive and often unexpected transcriptional response.
Collapse
Affiliation(s)
- Sonia Gandoura
- INSERM, U773, Centre de Recherche Biomédicale Bichat-Beaujon CRB3, Clichy and Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Islam MA, Pröll M, Hölker M, Tholen E, Tesfaye D, Looft C, Schellander K, Cinar MU. Alveolar macrophage phagocytic activity is enhanced with LPS priming, and combined stimulation of LPS and lipoteichoic acid synergistically induce pro-inflammatory cytokines in pigs. Innate Immun 2013; 19:631-43. [PMID: 23608822 DOI: 10.1177/1753425913477166] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The objective of the present study was to investigate LPS and lipoteichoic acid (LTA)-induced TLRs, associated signaling molecules and inflammatory mediators, as well as to compare their combined effect in porcine alveolar macrophages. Macrophages were incubated for 24 h with various concentrations of LPS, LTA, LPS + LTA or control. Multiple concentrations of LPS elicited marked up-regulation in mRNA for TLR2 and TLR4, CD14, MD2, MyD88, IRAK-4 and TRAF6 compared with the control. LTA had no effect on TLR4 and MD2; only higher doses up-regulated TLR2, CD14, MyD88, IRAK-4 and TRAF6 mRNA. LPS-activated cells released IL1-β, IL12-β, TNF-α, IL-6, IL-8, IFN-γ and IL-10 in a dose-dependent manner, while LTA had no effect on IL-1β, IL-6 and IFN-γ. Higher doses of LTA induced IL-12β, TNF-α, IL-8 and IL-10. Combined stimulation augmented TLR2, CD14 and MyD88 mRNA, and subsequently produced elevated levels of IL-6, TNF-α and IL-8 when compared with LPS and LTA alone. Additionally, phagocytosis of macrophages was significantly increased following low concentration of LPS treatment. Only low levels of NO (nitric oxide) were detected in the LPS group. Overall, compared with LPS, LTA was a relatively weak inducer, and co-stimulation accelerated gene and cytokine production associated with pulmonary innate immune function.
Collapse
|
69
|
Ahn SH, Tsalik EL, Cyr DD, Zhang Y, van Velkinburgh JC, Langley RJ, Glickman SW, Cairns CB, Zaas AK, Rivers EP, Otero RM, Veldman T, Kingsmore SF, Lucas J, Woods CW, Ginsburg GS, Fowler VG. Gene expression-based classifiers identify Staphylococcus aureus infection in mice and humans. PLoS One 2013; 8:e48979. [PMID: 23326304 PMCID: PMC3541361 DOI: 10.1371/journal.pone.0048979] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/27/2012] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus causes a spectrum of human infection. Diagnostic delays and uncertainty lead to treatment delays and inappropriate antibiotic use. A growing literature suggests the host’s inflammatory response to the pathogen represents a potential tool to improve upon current diagnostics. The hypothesis of this study is that the host responds differently to S. aureus than to E. coli infection in a quantifiable way, providing a new diagnostic avenue. This study uses Bayesian sparse factor modeling and penalized binary regression to define peripheral blood gene-expression classifiers of murine and human S. aureus infection. The murine-derived classifier distinguished S. aureus infection from healthy controls and Escherichia coli-infected mice across a range of conditions (mouse and bacterial strain, time post infection) and was validated in outbred mice (AUC>0.97). A S. aureus classifier derived from a cohort of 94 human subjects distinguished S. aureus blood stream infection (BSI) from healthy subjects (AUC 0.99) and E. coli BSI (AUC 0.84). Murine and human responses to S. aureus infection share common biological pathways, allowing the murine model to classify S. aureus BSI in humans (AUC 0.84). Both murine and human S. aureus classifiers were validated in an independent human cohort (AUC 0.95 and 0.92, respectively). The approach described here lends insight into the conserved and disparate pathways utilized by mice and humans in response to these infections. Furthermore, this study advances our understanding of S. aureus infection; the host response to it; and identifies new diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Sun Hee Ahn
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Ephraim L. Tsalik
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Section on Infectious Diseases, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
| | - Derek D. Cyr
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Yurong Zhang
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Jennifer C. van Velkinburgh
- van Velkinburgh Initiative for Collaborative BioMedical Research, Santa Fe, New Mexico, United States of America
| | - Raymond J. Langley
- Immunology Division, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States of America
| | - Seth W. Glickman
- Department of Emergency Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Charles B. Cairns
- Department of Emergency Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Aimee K. Zaas
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Emanuel P. Rivers
- Department of Emergency Medicine, Henry Ford Hospital, Wayne State University, Detroit, Michigan, United States of America
| | - Ronny M. Otero
- Department of Emergency Medicine, Henry Ford Hospital, Wayne State University, Detroit, Michigan, United States of America
| | - Tim Veldman
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Stephen F. Kingsmore
- Center for Pediatric Genomic Medicine, Children’s Mercy Hospitals and Clinics, Kansas City, Missouri, United States of America
| | - Joseph Lucas
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Section on Infectious Diseases, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Geoffrey S. Ginsburg
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
- * E-mail: (GSG); (VGF)
| | - Vance G. Fowler
- Division of Infectious Diseases and International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- * E-mail: (GSG); (VGF)
| |
Collapse
|
70
|
Gillmaier N, Götz A, Schulz A, Eisenreich W, Goebel W. Metabolic responses of primary and transformed cells to intracellular Listeria monocytogenes. PLoS One 2012; 7:e52378. [PMID: 23285016 PMCID: PMC3528701 DOI: 10.1371/journal.pone.0052378] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 11/12/2012] [Indexed: 02/08/2023] Open
Abstract
The metabolic response of host cells, in particular of primary mammalian cells, to bacterial infections is poorly understood. Here, we compare the carbon metabolism of primary mouse macrophages and of established J774A.1 cells upon Listeria monocytogenes infection using (13)C-labelled glucose or glutamine as carbon tracers. The (13)C-profiles of protein-derived amino acids from labelled host cells and intracellular L. monocytogenes identified active metabolic pathways in the different cell types. In the primary cells, infection with live L. monocytogenes increased glycolytic activity and enhanced flux of pyruvate into the TCA cycle via pyruvate dehydrogenase and pyruvate carboxylase, while in J774A.1 cells the already high glycolytic and glutaminolytic activities hardly changed upon infection. The carbon metabolism of intracellular L. monocytogenes was similar in both host cells. Taken together, the data suggest that efficient listerial replication in the cytosol of the host cells mainly depends on the glycolytic activity of the hosts.
Collapse
Affiliation(s)
- Nadine Gillmaier
- Lehrstuhl für Biochemie, Technische Universität München, Garching, Germany
| | - Andreas Götz
- Max-von-Pettenkofer Institut, Ludwig-Maximilians-Universität München, München, Germany
| | - Anette Schulz
- Max-von-Pettenkofer Institut, Ludwig-Maximilians-Universität München, München, Germany
| | | | - Werner Goebel
- Max-von-Pettenkofer Institut, Ludwig-Maximilians-Universität München, München, Germany
| |
Collapse
|
71
|
Francis SMS, Tan ME, Fung PR, Shaw JG, Semmler AB, Nataatmadja M, Bowman RV, Fong KM, Yang IA. Peripheral compartment innate immune response to Haemophilus influenzae and Streptococcus pneumoniae in chronic obstructive pulmonary disease patients. Innate Immun 2012; 19:428-37. [PMID: 23212542 DOI: 10.1177/1753425912466926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Alterations in innate immunity that predispose to chronic obstructive pulmonary disease (COPD) exacerbations are poorly understood. We examined innate immunity gene expression in peripheral blood polymorphonuclear leukocytes (PMN) and monocytes stimulated by Haemophilus influenzae and Streptococcus pneumoniae. Thirty COPD patients (15 rapid and 15 non-rapid lung function decliners) and 15 smokers without COPD were studied. Protein expression of IL-8, IL-6, TNF-α and IFN-γ (especially monocytes) increased with bacterial challenge. In monocytes stimulated with S. pneumoniae, TNF-α protein expression was higher in COPD (non-rapid decliners) than in smokers. In co-cultures of monocytes and PMN, mRNA expression of TGF-β1 and MYD88 was up-regulated, and CD14, TLR2 and IFN-γ down-regulated with H. influenzae challenge. TNF-α mRNA expression was increased with H. influenzae challenge in COPD. Cytokine responses were similar between rapid and non-rapid decliners. TNF-α expression was up-regulated in non-rapid decliners in response to H. influenzae (monocytes) and S. pneumoniae (co-culture of monocytes and PMN). Exposure to bacterial pathogens causes characteristic innate immune responses in peripheral blood monocytes and PMN in COPD. Bacterial exposure significantly alters the expression of TNF-α in COPD patients, although not consistently. There did not appear to be major differences in innate immune responses between rapid and non-rapid decliners.
Collapse
|
72
|
Tattermusch S, Bangham CRM. HTLV-1 infection: what determines the risk of inflammatory disease? Trends Microbiol 2012; 20:494-500. [PMID: 22917680 DOI: 10.1016/j.tim.2012.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 12/24/2022]
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is an exogenous retrovirus that persists lifelong in the infected host. Infection has been linked to a spectrum of diverse diseases: adult T cell leukemia, encephalomyelopathy, and predisposition to opportunistic bacterial and helminth infections. Applications of new technologies and biological concepts to the field have provided new insights into viral persistence and pathogenesis in HTLV-1 infection. Here, we summarize the emerging concepts of dynamic HTLV-1-host interactions and propose that chronic interferon (IFN) production causes tissue damage in HTLV-1-associated inflammatory diseases.
Collapse
Affiliation(s)
- Sonja Tattermusch
- Imperial College London, Department of Immunology, Norfolk Place, London W2 1PG, UK
| | | |
Collapse
|
73
|
Kyrova K, Stepanova H, Rychlik I, Faldyna M, Volf J. SPI-1 encoded genes of Salmonella Typhimurium influence differential polarization of porcine alveolar macrophages in vitro. BMC Vet Res 2012; 8:115. [PMID: 22817641 PMCID: PMC3441223 DOI: 10.1186/1746-6148-8-115] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/27/2012] [Indexed: 01/01/2023] Open
Abstract
Background Within the last decade, macrophages have been shown to be capable of differentiating toward a classically activated phenotype (M1) with a high antimicrobial potential or an alternatively activated phenotype (M2). Some pathogens are capable of interfering with differentiation in order to down-regulate the anti-microbial activity and enhance their survival in the host. Results To test this ability in Salmonella enterica serovar Typhimurium, we infected porcine alveolar macrophages with wild-type Salmonella Typhimurium and its isogenic mutants devoid of two major pathogenicity islands, SPI-1 and SPI-2. The induction of genes linked with M1 or M2 polarization was determined by quantification of gene expression by RT-qPCR. The ΔSPI-1 mutant induced a high, dose-dependent M1 response but a low M2 response in infected macrophages. On the other hand, wild-type Salmonella Typhimurium induced a low M1 response but a high, dose-dependent M2 response in infected macrophages. The response to ΔSPI-2 mutant infection was virtually the same as the wild-type strain. Conclusions We therefore propose that Salmonella Typhimurium DT104 studied here can polarize macrophages towards the less bactericidal M2 phenotype and that this polarization is dependent on the type III secretion system encoded by SPI-1.
Collapse
Affiliation(s)
- Kamila Kyrova
- Veterinary Research Institute, Hudcova Brno, Czech Republic
| | | | | | | | | |
Collapse
|
74
|
Wu K, Dong D, Fang H, Levillain F, Jin W, Mei J, Gicquel B, Du Y, Wang K, Gao Q, Neyrolles O, Zhang J. An interferon-related signature in the transcriptional core response of human macrophages to Mycobacterium tuberculosis infection. PLoS One 2012; 7:e38367. [PMID: 22675550 PMCID: PMC3366933 DOI: 10.1371/journal.pone.0038367] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 05/03/2012] [Indexed: 11/23/2022] Open
Abstract
The W-Beijing family of Mycobacterium tuberculosis (Mtb) strains is known for its high-prevalence and -virulence, as well as for its genetic diversity, as recently reported by our laboratories and others. However, little is known about how the immune system responds to these strains. To explore this issue, here we used reverse engineering and genome-wide expression profiling of human macrophage-like THP-1 cells infected by different Mtb strains of the W-Beijing family, as well as by the reference laboratory strain H37Rv. Detailed data mining revealed that host cell transcriptome responses to H37Rv and to different strains of the W-Beijing family are similar and overwhelmingly induced during Mtb infections, collectively typifying a robust gene expression signature (“THP1r2Mtb-induced signature”). Analysis of the putative transcription factor binding sites in promoter regions of genes in this signature identified several key regulators, namely STATs, IRF-1, IRF-7, and Oct-1, commonly involved in interferon-related immune responses. The THP1r2Mtb-induced signature appeared to be highly relevant to the interferon-inducible signature recently reported in active pulmonary tuberculosis patients, as revealed by cross-signature and cross-module comparisons. Further analysis of the publicly available transcriptome data from human patients showed that the signature appears to be relevant to active pulmonary tuberculosis patients and their clinical therapy, and be tuberculosis specific. Thus, our results provide an additional layer of information at the transcriptome level on mechanisms involved in host macrophage response to Mtb, which may also implicate the robustness of the cellular defense system that can effectively fight against genetic heterogeneity in this pathogen.
Collapse
Affiliation(s)
- Kang Wu
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Dong
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai Fang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florence Levillain
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
| | - Wen Jin
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Mei
- Department of Tuberculosis Control, Shanghai Municipal CDC, Shanghai, China
| | - Brigitte Gicquel
- Unité de Génétique Mycobactérienne, Institut Pasteur, Paris, France
| | - Yanzhi Du
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kankan Wang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Olivier Neyrolles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- * E-mail: (ON); (JZ)
| | - Ji Zhang
- State Key Laboratory of Medical Genomics and Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (ON); (JZ)
| |
Collapse
|
75
|
Lim A, Steibel JP, Coussens PM, Grooms DL, Bolin SR. Differential gene expression segregates cattle confirmed positive for bovine tuberculosis from antemortem tuberculosis test-false positive cattle originating from herds free of bovine tuberculosis. Vet Med Int 2012; 2012:192926. [PMID: 22701814 PMCID: PMC3373196 DOI: 10.1155/2012/192926] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 03/20/2012] [Accepted: 04/02/2012] [Indexed: 01/08/2023] Open
Abstract
Antemortem tests for bovine tuberculosis (bTB) currently used in the US measure cell-mediated immune responses against Mycobacterium bovis. Postmortem tests for bTB rely on observation of gross and histologic lesions of bTB, followed by bacterial isolation or molecular diagnostics. Cumulative data from the state of Michigan indicates that 98 to 99% of cattle that react positively in antemortem tests are not confirmed positive for bTB at postmortem examination. Understanding the fundamental differences in gene regulation between antemortem test-false positive cattle and cattle that have bTB may allow identification of molecular markers that can be exploited to better separate infected from noninfected cattle. An immunospecific cDNA microarray was used to identify altered gene expression (P ≤ 0.01) of 122 gene features between antemortem test-false positive cattle and bTB-infected cattle following a 4-hour stimulation of whole blood with tuberculin. Further analysis using quantitative real-time PCR assays validated altered expression of 8 genes that had differential power (adj P ≤ 0.05) to segregate cattle confirmed positive for bovine tuberculosis from antemortem tuberculosis test-false positive cattle originating from herds free of bovine tuberculosis.
Collapse
Affiliation(s)
- Ailam Lim
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
- Diagnostic Center for Population and Animal Health, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA
| | - Juan P. Steibel
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48824, USA
| | - Paul M. Coussens
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Daniel L. Grooms
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Steven R. Bolin
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
- Diagnostic Center for Population and Animal Health, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA
| |
Collapse
|
76
|
Tomioka H, Tatano Y, Maw WW, Sano C, Kanehiro Y, Shimizu T. Characteristics of suppressor macrophages induced by mycobacterial and protozoal infections in relation to alternatively activated M2 macrophages. Clin Dev Immunol 2012; 2012:635451. [PMID: 22666284 PMCID: PMC3361169 DOI: 10.1155/2012/635451] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 01/15/2023]
Abstract
In the advanced stages of mycobacterial infections, host immune systems tend to change from a Th1-type to Th2-type immune response, resulting in the abrogation of Th1 cell- and macrophage-mediated antimicrobial host protective immunity. Notably, this type of immune conversion is occasionally associated with the generation of certain types of suppressor macrophage populations. During the course of Mycobacterium tuberculosis (MTB) and Mycobacterium avium-intracellulare complex (MAC) infections, the generation of macrophages which possess strong suppressor activity against host T- and B-cell functions is frequently encountered. This paper describes the immunological properties of M1- and M2-type macrophages generated in tumor-bearing animals and those generated in hosts with certain microbial infections. In addition, this paper highlights the immunological and molecular biological characteristics of suppressor macrophages generated in hosts with mycobacterial infections, especially MAC infection.
Collapse
Affiliation(s)
- Haruaki Tomioka
- Department of Microbiology and Immunology, Shimane University School of Medicine, Izumo, Shimane 693-8501, Japan.
| | | | | | | | | | | |
Collapse
|
77
|
André MC, Gille C, Glemser P, Woiterski J, Hsu HY, Spring B, Keppeler H, Kramer BW, Handgretinger R, Poets CF, Lauber K, Orlikowsky TW. Bacterial reprogramming of PBMCs impairs monocyte phagocytosis and modulates adaptive T cell responses. J Leukoc Biol 2012; 91:977-89. [PMID: 22427683 DOI: 10.1189/jlb.0911474] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Septic diseases are characterized by an initial systemic, proinflammatory phase, followed by a period of anti-inflammation. In the context of the latter, monocytes have been described to display altered functions, including reduced TNF secretion and T cell-stimulating capacities in response to recall antigens. This hyporesponsiveness is supposed to be detrimental for coping with secondary infections. We here characterize bacterially reprogrammed PBMC-derived monocytes with special focus on their phagocytic activity. Hence, we have implemented a surrogate model of the early, postinflammatory period by exposing PBMCs to Escherichia coli on d0 and rechallenging them with bacteria on d2. This induced the emergence of a distinct monocytic phenotype with profound phagocytic impairments but a preserved ability for naïve T cell stimulation. The compromising effects on phagocytosis required the presence of bacteria and were not mimicked by TLR4 ligation or exposure to isolated cytokines alone. Moreover, the impairments were specific for the engulfment of bacteria and were coupled to a selective down-regulation of FcγR and SR expression. Intriguingly, this monocytic phenotype contributed to the stimulation of a T(H)17-polarized adaptive immune response in the context of secondary infection. Our findings extend the current knowledge of monocytic reprogramming and identify the phagocytic capacity of monocytes as a putative sepsis biomarker.
Collapse
Affiliation(s)
- Maya C André
- Department of Neonatology, University Children’s Hospital, Tuebingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Bonnefont CMD, Rainard P, Cunha P, Gilbert FB, Toufeer M, Aurel MR, Rupp R, Foucras G. Genetic susceptibility to S. aureus mastitis in sheep: differential expression of mammary epithelial cells in response to live bacteria or supernatant. Physiol Genomics 2012; 44:403-16. [PMID: 22337903 DOI: 10.1152/physiolgenomics.00155.2011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Staphylococcus aureus is a prevalent pathogen for mastitis in dairy ruminants and is responsible for both clinical and subclinical mastitis. Mammary epithelial cells (MEC) represent not only a physical barrier against bacterial invasion but are also active players of the innate immune response permitting infection clearance. To decipher their functions in general and in animals showing different levels of genetic predisposition to Staphylococcus in particular, MEC from ewes undergoing a divergent selection on milk somatic cell count were stimulated by S. aureus. MEC response was also studied according to the stimulation condition with live bacteria or culture supernatant. The early MEC response was studied during a 5 h time course by microarray to identify differentially expressed genes with regard to the host genetic background and as a function of the conditions of stimulation. In both conditions of stimulation, metabolic processes were altered, the apoptosis-associated pathways were considerably modified, and inflammatory and immune responses were enhanced with the upregulation of il1a, il1b, and tnfa and several chemokines known to enhance neutrophil (cxcl8) or mononuclear leukocyte (ccl20) recruitment. Genes associated with oxidative stress were increased after live bacteria stimulation, whereas immune response-related genes were higher after supernatant stimulation in the early phase. Only 20 genes were differentially expressed between Staphylococcus spp-mastitis resistant and susceptible animals without any clearly defined role on the control of infection. To conclude, this suggests that MEC may not represent the cell type at the origin of the difference of mastitis susceptibility, at least as demonstrated in our genetic model. Supernatant or heat-killed S. aureus produce biological effects that are essentially different from those induced by live bacteria.
Collapse
Affiliation(s)
- Cécile M D Bonnefont
- Université de Toulouse, Institut National Polytechnique (INP), École Nationale Vétérinaire de Toulouse (ENVT), Unité Mixte de Recherche (UMR)1225, Interactions Hôtes - Agents Pathogènes (IHAP), Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Kim HS. Our Genome and Our other Genome: Understanding humans as Symbionts with Microbes. ACTA ACUST UNITED AC 2012. [DOI: 10.4167/jbv.2012.42.2.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Heenam Stanley Kim
- The Laboratory of Human-Microbial Genomics, Department of Medicine, College of Medicine, Korea University, Anam-Dong, Seongbuk-Gu, Seoul, Korea
| |
Collapse
|
80
|
Debey-Pascher S, Chen J, Voss T, Staratschek-Jox A. Blood-based miRNA preparation for noninvasive biomarker development. Methods Mol Biol 2012; 822:307-38. [PMID: 22144209 DOI: 10.1007/978-1-61779-427-8_22] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This chapter describes several methods for the isolation of miRNAs from peripheral whole blood samples or constituent fractions thereof, such as peripheral blood mononuclear cells, plasma, and serum. The methods described here are recently introduced protocols dedicated to the isolation of total RNAs including small RNAs, e.g., miRNeasy Kit and PAXgene Blood miRNA Kit, or alternatively for the enrichment of low-molecular-weight RNA (LMW RNA) fractions including small RNAs, e.g., using the miRNeasy Kit. Furthermore, modifications of classical RNA purification protocols to facilitate the recovery of small RNAs are highlighted.
Collapse
Affiliation(s)
- Svenja Debey-Pascher
- Life and Medical Sciences (LIMES), Genomics and Immunoregulation, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|
81
|
Yen JY, Garamszegi S, Geisbert JB, Rubins KH, Geisbert TW, Honko A, Xia Y, Connor JH, Hensley LE. Therapeutics of Ebola hemorrhagic fever: whole-genome transcriptional analysis of successful disease mitigation. J Infect Dis 2011; 204 Suppl 3:S1043-52. [PMID: 21987740 DOI: 10.1093/infdis/jir345] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The mechanisms of Ebola (EBOV) pathogenesis are only partially understood, but the dysregulation of normal host immune responses (including destruction of lymphocytes, increases in circulating cytokine levels, and development of coagulation abnormalities) is thought to play a major role. Accumulating evidence suggests that much of the observed pathology is not the direct result of virus-induced structural damage but rather is due to the release of soluble immune mediators from EBOV-infected cells. It is therefore essential to understand how the candidate therapeutic may be interrupting the disease process and/or targeting the infectious agent. To identify genetic signatures that are correlates of protection, we used a DNA microarray-based approach to compare the host genome-wide responses of EBOV-infected nonhuman primates (NHPs) responding to candidate therapeutics. We observed that, although the overall circulating immune response was similar in the presence and absence of coagulation inhibitors, surviving NHPs clustered together. Noticeable differences in coagulation-associated genes appeared to correlate with survival, which revealed a subset of distinctly differentially expressed genes, including chemokine ligand 8 (CCL8/MCP-2), that may provide possible targets for early-stage diagnostics or future therapeutics. These analyses will assist us in understanding the pathogenic mechanisms of EBOV infection and in identifying improved therapeutic strategies.
Collapse
Affiliation(s)
- Judy Y Yen
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Modulation of the host interferon response and ISGylation pathway by B. pertussis filamentous hemagglutinin. PLoS One 2011; 6:e27535. [PMID: 22140447 PMCID: PMC3227562 DOI: 10.1371/journal.pone.0027535] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 10/19/2011] [Indexed: 11/19/2022] Open
Abstract
Bordetella pertussis filamentous hemagglutinin (FHA) is a surface-associated and secreted protein that serves as a crucial adherence factor, and displays immunomodulatory activity in human peripheral blood mononuclear cells (PBMCs). In order to appreciate more fully the role of secreted FHA in pathogenesis, we analyzed FHA-induced changes in genome-wide transcript abundance in human PBMCs. Among the 683 known unique genes with greater than 3-fold change in transcript abundance following FHA treatment, 125 (18.3%) were identified as interferon (IFN)-regulated. Among the latter group were genes encoding several members of the IFN type I response, as well as 3 key components of the ISGylation pathway. Using real-time RT-PCR, we confirmed FHA-associated increases in transcript abundance for the genes encoding ubiquitin-like protein, ISG15, and its specific protease USP18. Western-blot analysis demonstrated the presence of both, free ISG15 and several ISGylated conjugates in FHA-stimulated PBMC lysates, but not in unstimulated cells. Intracellular FACS analysis provided evidence that monocytes and a natural killer-enriched cell population were the primary producers of ISG15 in PBMCs after FHA stimulation. Our data reveal previously-unrecognized effects of B. pertussis FHA on host IFN and ISGylation responses, and suggest previously-unsuspected mechanisms by which FHA may alter the outcome of the host-pathogen interaction.
Collapse
|
83
|
Joehanes R, Johnson AD, Barb JJ, Raghavachari N, Liu P, Woodhouse KA, O'Donnell CJ, Munson PJ, Levy D. Gene expression analysis of whole blood, peripheral blood mononuclear cells, and lymphoblastoid cell lines from the Framingham Heart Study. Physiol Genomics 2011; 44:59-75. [PMID: 22045913 DOI: 10.1152/physiolgenomics.00130.2011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Despite a growing number of reports of gene expression analysis from blood-derived RNA sources, there have been few systematic comparisons of various RNA sources in transcriptomic analysis or for biomarker discovery in the context of cardiovascular disease (CVD). As a pilot study of the Systems Approach to Biomarker Research (SABRe) in CVD Initiative, this investigation used Affymetrix Exon arrays to characterize gene expression of three blood-derived RNA sources: lymphoblastoid cell lines (LCL), whole blood using PAXgene tubes (PAX), and peripheral blood mononuclear cells (PBMC). Their performance was compared in relation to identifying transcript associations with sex and CVD risk factors, such as age, high-density lipoprotein, and smoking status, and the differential blood cell count. We also identified a set of exons that vary substantially between participants, but consistently in each RNA source. Such exons are thus stable phenotypes of the participant and may potentially become useful fingerprinting biomarkers. In agreement with previous studies, we found that each of the RNA sources is distinct. Unlike PAX and PBMC, LCL gene expression showed little association with the differential blood count. LCL, however, was able to detect two genes related to smoking status. PAX and PBMC identified Y-chromosome probe sets similarly and slightly better than LCL.
Collapse
Affiliation(s)
- Roby Joehanes
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Evans SE, Tuvim MJ, Fox CJ, Sachdev N, Gibiansky L, Dickey BF. Inhaled innate immune ligands to prevent pneumonia. Br J Pharmacol 2011; 163:195-206. [PMID: 21250981 DOI: 10.1111/j.1476-5381.2011.01237.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epithelial surfaces throughout the body continuously sample and respond to environmental stimuli. The accessibility of lung epithelium to inhaled therapies makes it possible to stimulate local antimicrobial defences with aerosolized innate immune ligands. This strategy has been shown to be effective in preclinical models, as delivery of innate immune ligands to the lungs of laboratory animals results in protection from subsequent challenge with microbial pathogens. Survival of the animal host in this setting correlates directly with killing of pathogens within the lungs, indicating the induction of a resistance mechanism. Resistance appears to be mediated primarily by activated epithelial cells rather than recruited leucocytes. Resistance reaches a peak within hours and persists for several days. Innate immune ligands can interact synergistically under some circumstances, and synergistic combinations of innate ligands delivered by aerosol are capable of inducing a high level of broad host resistance to bacteria, fungi and viruses. The induction of innate antimicrobial resistance within the lungs could have clinical applications in the prevention of lower respiratory tract infection in subjects transiently at high risk. These include cancer patients undergoing myeloablative chemotherapy, intubated patients being mechanically ventilated, vulnerable individuals during seasonal influenza epidemics, asthmatic subjects experiencing a respiratory viral infection, and healthy subjects exposed to virulent pathogens from a bioterror attack or emergent pandemic. In summary, stimulation of the lung epithelium to induce localized resistance to infection is a novel strategy whose clinical utility will be assessed in the near future.
Collapse
Affiliation(s)
- Scott E Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | | | | | | | | | | |
Collapse
|
85
|
Transcriptomic and nuclear architecture of immune cells after LPS activation. Chromosoma 2011; 120:501-20. [DOI: 10.1007/s00412-011-0328-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 06/01/2011] [Accepted: 06/01/2011] [Indexed: 01/06/2023]
|
86
|
Germain RN, Meier-Schellersheim M, Nita-Lazar A, Fraser IDC. Systems biology in immunology: a computational modeling perspective. Annu Rev Immunol 2011; 29:527-85. [PMID: 21219182 DOI: 10.1146/annurev-immunol-030409-101317] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Systems biology is an emerging discipline that combines high-content, multiplexed measurements with informatic and computational modeling methods to better understand biological function at various scales. Here we present a detailed review of the methods used to create computational models and to conduct simulations of immune function. We provide descriptions of the key data-gathering techniques employed to generate the quantitative and qualitative data required for such modeling and simulation and summarize the progress to date in applying these tools and techniques to questions of immunological interest, including infectious disease. We include comments on what insights modeling can provide that complement information obtained from the more familiar experimental discovery methods used by most investigators and the reasons why quantitative methods are needed to eventually produce a better understanding of immune system operation in health and disease.
Collapse
Affiliation(s)
- Ronald N Germain
- Program in Systems Immunology and Infectious Disease Modeling, National Institute of Allergy and Infectious Disease, Laboratory of Immunology, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
87
|
Heath RJW, Leong JM, Visegrády B, Machesky LM, Xavier RJ. Bacterial and host determinants of MAL activation upon EPEC infection: the roles of Tir, ABRA, and FLRT3. PLoS Pathog 2011; 7:e1001332. [PMID: 21490959 PMCID: PMC3072376 DOI: 10.1371/journal.ppat.1001332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 03/10/2011] [Indexed: 11/23/2022] Open
Abstract
Infection of host cells by pathogenic microbes triggers signal transduction pathways leading to a multitude of host cell responses including actin cytoskeletal re-arrangements and transcriptional programs. The diarrheagenic pathogens Enteropathogenic E. coli (EPEC) and the related Enterohemorrhagic E. coli (EHEC) subvert the host-cell actin cytoskeleton to form attaching and effacing lesions on the surface of intestinal epithelial cells by injecting effector proteins via a type III secretion system. Here we use a MAL translocation assay to establish the effect of bacterial pathogens on host cell signaling to transcription factor activation. MAL is a cofactor of Serum response factor (SRF), a transcription factor with important roles in the regulation of the actin cytoskeleton. We show that EPEC induces nuclear accumulation of MAL-GFP. The translocated intimin receptor is essential for this process and phosphorylation of Tyrosine residues 454 and 474 is important. Using an expression screen we identify FLRT3, C22orf28 and TESK1 as novel activators of SRF. Importantly we demonstrate that ABRA (actin-binding Rho-activating protein, also known as STARS) is necessary for EPEC-induced nuclear accumulation of MAL and the novel SRF activator FLRT3, is a component of this pathway. We further demonstrate that ABRA is important for structural maintenance of EPEC pedestals. Our results uncover novel components in pathogen-activated cytoskeleton signalling to MAL activation. Many significant immune diseases are caused by bacterial pathogens that deliver effector proteins into their host. The pathogen uses these proteins to subvert the hosts' normal cytosolic defense in a way that services the pathogen. It is therefore important to understand the normal processes of a cell and how they are affected by bacterial infection. We have established the effect of bacteria on host cell signalling to the transcription factor serum response factor. Serum response factor is a widely expressed transcription factor that controls the expression of many important genes. We show that Enteropathogenic E. coli infection can activate serum response factor and that the effector protein Tir is essential for this activation. Furthermore, we identify new genes that are important in this infection-induced activation and show that they are important in maintaining structures necessary for Enteropathogenic E. coli infection.
Collapse
Affiliation(s)
- Robert J. W. Heath
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Gastrointestinal Unit, Center for Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - John M. Leong
- Department of Molecular Genetics and Microbiology, UMass Medical School, Worcester, Massachusetts, United States of America
| | - Balázs Visegrády
- Beatson Institute for Cancer Research, Garscube Estate, Bearsden, Glasgow, United Kingdom
| | - Laura M. Machesky
- Beatson Institute for Cancer Research, Garscube Estate, Bearsden, Glasgow, United Kingdom
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Gastrointestinal Unit, Center for Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
88
|
Rubins KH, Hensley LE, Relman DA, Brown PO. Stunned silence: gene expression programs in human cells infected with monkeypox or vaccinia virus. PLoS One 2011; 6:e15615. [PMID: 21267444 PMCID: PMC3022624 DOI: 10.1371/journal.pone.0015615] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/12/2010] [Indexed: 12/20/2022] Open
Abstract
Poxviruses use an arsenal of molecular weapons to evade detection and disarm host immune responses. We used DNA microarrays to investigate the gene expression responses to infection by monkeypox virus (MPV), an emerging human pathogen, and Vaccinia virus (VAC), a widely used model and vaccine organism, in primary human macrophages, primary human fibroblasts and HeLa cells. Even as the overwhelmingly infected cells approached their demise, with extensive cytopathic changes, their gene expression programs appeared almost oblivious to poxvirus infection. Although killed (gamma-irradiated) MPV potently induced a transcriptional program characteristic of the interferon response, no such response was observed during infection with either live MPV or VAC. Moreover, while the gene expression response of infected cells to stimulation with ionomycin plus phorbol 12-myristate 13-acetate (PMA), or poly (I-C) was largely unimpaired by infection with MPV, a cluster of pro-inflammatory genes were a notable exception. Poly(I-C) induction of genes involved in alerting the innate immune system to the infectious threat, including TNF-alpha, IL-1 alpha and beta, CCL5 and IL-6, were suppressed by infection with live MPV. Thus, MPV selectively inhibits expression of genes with critical roles in cell-signaling pathways that activate innate immune responses, as part of its strategy for stealthy infection.
Collapse
Affiliation(s)
- Kathleen H. Rubins
- Departments of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lisa E. Hensley
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - David A. Relman
- Departments of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Patrick O. Brown
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
89
|
Abstract
Recent technological developments have revolutionized our understanding of transcriptional regulation by providing an unprecedented ability to interrogate in vivo transcription factor binding. The combination of high-throughput sequencing with chromatin precipitation of transcription factors and specifically labeled histones has allowed direct protein-DNA contacts to be visualized across genomes as large and complex as mammals at base-pair resolution. This chapter reviews the developments that led to these insights, with particular focus on examples of early protein-DNA localization experiments using genomic microarrays in mammals and yeast. Four state-of-the-art research directions are highlighted as examples of previously unimaginable frontiers now under active investigation.
Collapse
Affiliation(s)
- Duncan T Odom
- Cancer Research UK, Li Ka Shing Centre, University of Cambridge, Cambridge, UK,
| |
Collapse
|
90
|
Chin CY, Monack DM, Nathan S. Genome wide transcriptome profiling of a murine acute melioidosis model reveals new insights into how Burkholderia pseudomallei overcomes host innate immunity. BMC Genomics 2010; 11:672. [PMID: 21110886 PMCID: PMC3017868 DOI: 10.1186/1471-2164-11-672] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 11/27/2010] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND At present, very little is known about how Burkholderia pseudomallei (B. pseudomallei) interacts with its host to elicit melioidosis symptoms. We established a murine acute-phase melioidosis model and used DNA microarray technology to investigate the global host/pathogen interaction. We compared the transcriptome of infected liver and spleen with uninfected tissues over an infection period of 42 hr to identify genes whose expression is altered in response to an acute infection. RESULTS Viable B. pseudomallei cells were consistently detected in the blood, liver and spleen during the 42 hr course of infection. Microarray analysis of the liver and spleen over this time course demonstrated that genes involved in immune response, stress response, cell cycle regulation, proteasomal degradation, cellular metabolism and signal transduction pathways were differentially regulated. Up regulation of toll-like receptor 2 (TLR2) gene expression suggested that a TLR2-mediated signalling pathway is responsible for recognition and initiation of an inflammatory response to the acute B. pseudomallei infection. Most of the highly elevated inflammatory genes are a cohort of "core host immune response" genes commonly seen in general inflammation infections. Concomitant to this initial inflammatory response, we observed an increase in transcripts associated with cell-death, caspase activation and peptidoglysis that ultimately promote tissue injury in the host. The complement system responsible for restoring host cellular homeostasis and eliminating intracellular bacteria was activated only after 24 hr post-infection. However, at this time point, diverse host nutrient metabolic and cellular pathways including glycolysis, fatty acid metabolism and tricarboxylic acid (TCA) cycle were repressed. CONCLUSIONS This detailed picture of the host transcriptional response during acute melioidosis highlights a broad range of innate immune mechanisms that are activated in the host within 24 hrs, including the core immune response commonly seen in general inflammatory infections. Nevertheless, this activation is suppressed at 42 hr post-infection and in addition, suboptimal activation and function of the downstream complement system promotes uncontrolled spread of the bacteria.
Collapse
Affiliation(s)
- Chui-Yoke Chin
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi Selangor D. E. Malaysia
| | | | | |
Collapse
|
91
|
|
92
|
Transcriptomic and innate immune responses to Yersinia pestis in the lymph node during bubonic plague. Infect Immun 2010; 78:5086-98. [PMID: 20876291 DOI: 10.1128/iai.00256-10] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
A delayed inflammatory response is a prominent feature of infection with Yersinia pestis, the agent of bubonic and pneumonic plague. Using a rat model of bubonic plague, we examined lymph node histopathology, transcriptome, and extracellular cytokine levels to broadly characterize the kinetics and extent of the host response to Y. pestis and how it is influenced by the Yersinia virulence plasmid (pYV). Remarkably, dissemination and multiplication of wild-type Y. pestis during the bubonic stage of disease did not induce any detectable gene expression or cytokine response by host lymph node cells in the developing bubo. Only after systemic spread had led to terminal septicemic plague was a transcriptomic response detected, which included upregulation of several cytokine, chemokine, and other immune response genes. Although an initial intracellular phase of Y. pestis infection has been postulated, a Th1-type cytokine response associated with classical activation of macrophages was not observed during the bubonic stage of disease. However, elevated levels of interleukin-17 (IL-17) were present in infected lymph nodes. In the absence of pYV, sustained recruitment to the lymph node of polymorphonuclear leukocytes (PMN, or neutrophils), the major IL-17 effector cells, correlated with clearance of infection. Thus, the ability to counteract a PMN response in the lymph node appears to be a major in vivo function of the Y. pestis virulence plasmid.
Collapse
|
93
|
Zaas AK, Aziz H, Lucas J, Perfect JR, Ginsburg GS. Blood gene expression signatures predict invasive candidiasis. Sci Transl Med 2010; 2:21ra17. [PMID: 20374997 DOI: 10.1126/scitranslmed.3000715] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Candidemia is the fourth most common bloodstream infection, with Candida albicans being the most common causative species. Success in reducing the associated morbidity and mortality has been limited by the inadequacy and time delay of currently available diagnostic modalities. Focusing on host response to infection, we used a murine model to develop a blood gene expression signature that accurately classified mice with candidemia and distinguished candidemia from Staphylococcus aureus bacteremia. Validation of the signature was achieved in an independent cohort of mice. Genes represented in the signature have known associations with host defense against Candida and other microorganisms. Our results demonstrate a temporal pattern of host molecular responses that distinguish candidemia from S. aureus-induced bacteremia and establish a novel paradigm for infectious disease diagnosis.
Collapse
Affiliation(s)
- Aimee K Zaas
- Institute for Genome Sciences and Policy, Duke University, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
94
|
Evans SE, Tuvim MJ, Zhang J, Larson DT, García CD, Martinez-Pro S, Coombes KR, Dickey BF. Host lung gene expression patterns predict infectious etiology in a mouse model of pneumonia. Respir Res 2010; 11:101. [PMID: 20653947 PMCID: PMC2914038 DOI: 10.1186/1465-9921-11-101] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 07/23/2010] [Indexed: 11/28/2022] Open
Abstract
Background Lower respiratory tract infections continue to exact unacceptable worldwide mortality, often because the infecting pathogen cannot be identified. The respiratory epithelia provide protection from pneumonias through organism-specific generation of antimicrobial products, offering potential insight into the identity of infecting pathogens. This study assesses the capacity of the host gene expression response to infection to predict the presence and identity of lower respiratory pathogens without reliance on culture data. Methods Mice were inhalationally challenged with S. pneumoniae, P. aeruginosa, A. fumigatus or saline prior to whole genome gene expression microarray analysis of their pulmonary parenchyma. Characteristic gene expression patterns for each condition were identified, allowing the derivation of prediction rules for each pathogen. After confirming the predictive capacity of gene expression data in blinded challenges, a computerized algorithm was devised to predict the infectious conditions of subsequent subjects. Results We observed robust, pathogen-specific gene expression patterns as early as 2 h after infection. Use of an algorithmic decision tree revealed 94.4% diagnostic accuracy when discerning the presence of bacterial infection. The model subsequently differentiated between bacterial pathogens with 71.4% accuracy and between non-bacterial conditions with 70.0% accuracy, both far exceeding the expected diagnostic yield of standard culture-based bronchoscopy with bronchoalveolar lavage. Conclusions These data substantiate the specificity of the pulmonary innate immune response and support the feasibility of a gene expression-based clinical tool for pneumonia diagnosis.
Collapse
Affiliation(s)
- Scott E Evans
- Department of Pulmonary Medicine, University of Texas-M D, Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Callegan MC. Checks and balances: the ocular response to infection. Virulence 2010; 1:222. [PMID: 21178447 DOI: 10.4161/viru.1.4.12317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Bacterial corneal infections threaten vision. With the widespread use of contact lenses and the increasing number of vision-correction (refractive) surgeries, the number of bacterial corneal infection (keratitis) cases has dramatically increased over the past decade. These infections are often blinding, as bacteria multiply in the corneal epithelium and stroma, provoking inflammatory cell migration into the cornea, and ultimately damage or destruction of corneal tissue.
Collapse
|
96
|
Heimer SR, Yamada A, Russell H, Gilmore M. Response of corneal epithelial cells to Staphylococcus aureus. Virulence 2010; 1:223-35. [PMID: 21178448 PMCID: PMC3073293 DOI: 10.4161/viru.1.4.11466] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 02/04/2010] [Accepted: 02/08/2010] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus is a leading cause of invasive infection. It also infects wet mucosal tissues including the cornea and conjunctiva. Conflicting evidence exists on the expression of Toll-like receptors by human corneal epithelial cells. It was therefore of interest to determine how epithelial cells from this immune privileged tissue respond to S. aureus. Further, it was of interest to determine whether cytolytic toxins, with the potential to cause ion flux or potentially permit effector molecule movement across the target cell membrane, alter the response. Microarrays were used to globally assess the response of human corneal epithelial cells to S. aureus. A large increase in abundance of transcripts encoding the antimicrobial dendritic cell chemokine, CCL20, was observed. CCL20 release into the medium was detected, and this response was found to be largely TLR2 and NOD2 independent. Corneal epithelial cells also respond to S. aureus by increasing the intracellular abundance of mRNA for inflammatory mediators, transcription factors, and genes related to MAP kinase pathways, in ways similar to other cell types. The corneal epithelial cell response was surprisingly unaffected by toxin exposure. Toxin exposure did, however, induce a stress response. Although model toxigenic and non-toxigenic strains of S. aureus were employed in the present study, the results obtained were strikingly similar to those reported for stimulation of vaginal epithelial cells by clinical toxic shock toxin expressing isolates, demonstrating that the initial epithelial cellular responses to S. aureus are largely independent of strain as well as epithelial cell tissue source.
Collapse
|
97
|
Peng SC, Wong DSH, Tung KC, Chen YY, Chao CC, Peng CH, Chuang YJ, Tang CY. Computational modeling with forward and reverse engineering links signaling network and genomic regulatory responses: NF-kappaB signaling-induced gene expression responses in inflammation. BMC Bioinformatics 2010; 11:308. [PMID: 20529327 PMCID: PMC2889938 DOI: 10.1186/1471-2105-11-308] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 06/08/2010] [Indexed: 11/30/2022] Open
Abstract
Background Signal transduction is the major mechanism through which cells transmit external stimuli to evoke intracellular biochemical responses. Diverse cellular stimuli create a wide variety of transcription factor activities through signal transduction pathways, resulting in different gene expression patterns. Understanding the relationship between external stimuli and the corresponding cellular responses, as well as the subsequent effects on downstream genes, is a major challenge in systems biology. Thus, a systematic approach is needed to integrate experimental data and theoretical hypotheses to identify the physiological consequences of environmental stimuli. Results We proposed a systematic approach that combines forward and reverse engineering to link the signal transduction cascade with the gene responses. To demonstrate the feasibility of our strategy, we focused on linking the NF-κB signaling pathway with the inflammatory gene regulatory responses because NF-κB has long been recognized to play a crucial role in inflammation. We first utilized forward engineering (Hybrid Functional Petri Nets) to construct the NF-κB signaling pathway and reverse engineering (Network Components Analysis) to build a gene regulatory network (GRN). Then, we demonstrated that the corresponding IKK profiles can be identified in the GRN and are consistent with the experimental validation of the IKK kinase assay. We found that the time-lapse gene expression of several cytokines and chemokines (TNF-α, IL-1, IL-6, CXCL1, CXCL2 and CCL3) is concordant with the NF-κB activity profile, and these genes have stronger influence strength within the GRN. Such regulatory effects have highlighted the crucial roles of NF-κB signaling in the acute inflammatory response and enhance our understanding of the systemic inflammatory response syndrome. Conclusion We successfully identified and distinguished the corresponding signaling profiles among three microarray datasets with different stimuli strengths. In our model, the crucial genes of the NF-κB regulatory network were also identified to reflect the biological consequences of inflammation. With the experimental validation, our strategy is thus an effective solution to decipher cross-talk effects when attempting to integrate new kinetic parameters from other signal transduction pathways. The strategy also provides new insight for systems biology modeling to link any signal transduction pathways with the responses of downstream genes of interest.
Collapse
Affiliation(s)
- Shih Chi Peng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Gao Y, Flori L, Lecardonnel J, Esquerré D, Hu ZL, Teillaud A, Lemonnier G, Lefèvre F, Oswald IP, Rogel-Gaillard C. Transcriptome analysis of porcine PBMCs after in vitro stimulation by LPS or PMA/ionomycin using an expression array targeting the pig immune response. BMC Genomics 2010; 11:292. [PMID: 20459780 PMCID: PMC2881026 DOI: 10.1186/1471-2164-11-292] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 05/11/2010] [Indexed: 01/05/2023] Open
Abstract
Background Designing sustainable animal production systems that better balance productivity and resistance to disease is a major concern. In order to address questions related to immunity and resistance to disease in pig, it is necessary to increase knowledge on its immune system and to produce efficient tools dedicated to this species. Results A long-oligonucleotide-based chip referred to as SLA-RI/NRSP8-13K was produced by combining a generic set with a newly designed SLA-RI set that targets all annotated loci of the pig major histocompatibility complex (MHC) region (SLA complex) in both orientations as well as immunity genes outside the SLA complex. The chip was used to study the immune response of pigs following stimulation of porcine peripheral blood mononuclear cells (PBMCs) with lipopolysaccharide (LPS) or a mixture of phorbol myristate acetate (PMA) and ionomycin for 24 hours. Transcriptome analysis revealed that ten times more genes were differentially expressed after PMA/ionomycin stimulation than after LPS stimulation. LPS stimulation induced a general inflammation response with over-expression of SAA1, pro-inflammatory chemokines IL8, CCL2, CXCL5, CXCL3, CXCL2 and CCL8 as well as genes related to oxidative processes (SOD2) and calcium pathways (S100A9 and S100A12). PMA/ionomycin stimulation induced a stronger up-regulation of T cell activation than of B cell activation with dominance toward a Th1 response, including IL2, CD69 and TNFRSF9 (tumor necrosis factor receptor superfamily, member 9) genes. In addition, a very intense repression of THBS1 (thrombospondin 1) was observed. Repression of MHC class I genes was observed after PMA/ionomycin stimulation despite an up-regulation of the gene cascade involved in peptide processing. Repression of MHC class II genes was observed after both stimulations. Our results provide preliminary data suggesting that antisense transcripts mapping to the SLA complex may have a role during immune response. Conclusion The SLA-RI/NRSP8-13K chip was found to accurately decipher two distinct immune response activations of PBMCs indicating that it constitutes a valuable tool to further study immunity and resistance to disease in pig. The transcriptome analysis revealed specific and common features of the immune responses depending on the stimulation agent that increase knowledge on pig immunity.
Collapse
Affiliation(s)
- Yu Gao
- INRA, UMR 1313 de Génétique Animale et Biologie Intégrative, Domaine de Vilvert, 78350 Jouy-en-Josas, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Pascual V, Chaussabel D, Banchereau J. A genomic approach to human autoimmune diseases. Annu Rev Immunol 2010; 28:535-71. [PMID: 20192809 DOI: 10.1146/annurev-immunol-030409-101221] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The past decade has seen an explosion in the use of DNA-based microarrays. These techniques permit assessment of RNA abundance on a genome-wide scale. Medical applications emerged in the field of cancer, with studies of both solid tumors and hematological malignancies leading to the development of tests that are now used to personalize therapeutic options. Microarrays have also been used to analyze the blood transcriptome in a wide range of diseases. In human autoimmune diseases, these studies are showing potential for identifying therapeutic targets as well as biomarkers for diagnosis, assessment of disease activity, and response to treatment. More quantitative and sensitive high-throughput RNA profiling methods are starting to be available and will be necessary for transcriptome analyses to become routine tests in the clinical setting. We expect this to crystallize within the coming decade, as these methods become part of the personalized medicine armamentarium.
Collapse
Affiliation(s)
- Virginia Pascual
- Baylor Institute for Immunology Research, INSERM U, Dallas, Texas 75204, USA.
| | | | | |
Collapse
|
100
|
Evans SE, Xu Y, Tuvim MJ, Dickey BF. Inducible innate resistance of lung epithelium to infection. Annu Rev Physiol 2010; 72:413-35. [PMID: 20148683 DOI: 10.1146/annurev-physiol-021909-135909] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Most studies of innate immunity have focused on leukocytes such as neutrophils, macrophages, and natural killer cells. However, epithelial cells play key roles in innate defenses that include providing a mechanical barrier to microbial entry, signaling to leukocytes, and directly killing pathogens. Importantly, all these defenses are highly inducible in response to the sensing of microbial and host products. In healthy lungs, the level of innate immune epithelial function is low at baseline. This is indicated by low levels of spontaneous microbial killing and cytokine release, reflecting low constitutive stimulation in the nearly sterile lower respiratory tract when mucociliary clearance mechanisms are functioning effectively. This contrasts with the colon, where bacteria are continuously present and epithelial cells are constitutively activated. Although the surface area of the lungs presents a large target for microbial invasion, activated lung epithelial cells that are closely apposed to deposited pathogens are ideally positioned for microbial killing.
Collapse
Affiliation(s)
- Scott E Evans
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|