51
|
Abdelaziz MO, Ossmann S, Kaufmann AM, Leitner J, Steinberger P, Willimsky G, Raftery MJ, Schönrich G. Development of a Human Cytomegalovirus (HCMV)-Based Therapeutic Cancer Vaccine Uncovers a Previously Unsuspected Viral Block of MHC Class I Antigen Presentation. Front Immunol 2019; 10:1776. [PMID: 31417555 PMCID: PMC6682651 DOI: 10.3389/fimmu.2019.01776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) induces a uniquely high frequency of virus-specific effector/memory CD8+ T-cells, a phenomenon termed “memory inflation”. Thus, HCMV-based vaccines are particularly interesting in order to stimulate a sustained and strong cellular immune response against cancer. Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with high lethality and inevitable relapse. The current standard treatment does not significantly improve the desperate situation underlining the urgent need to develop novel approaches. Although HCMV is highly fastidious with regard to species and cell type, GBM cell lines are susceptible to HCMV. In order to generate HCMV-based therapeutic vaccine candidates, we deleted all HCMV-encoded proteins (immunoevasins) that interfere with MHC class I presentation. The aim being to use the viral vector as an adjuvant for presentation of endogenous tumor antigens, the presentation of high levels of vector-encoded neoantigens and finally the repurposing of bystander HCMV-specific CD8+ T cells to fight the tumor. As neoantigen, we exemplarily used the E6 and E7 proteins of human papillomavirus type 16 (HPV-16) as a non-transforming fusion protein (E6/E7) that covers all relevant antigenic peptides. Surprisingly, GBM cells infected with E6/E7-expressing HCMV-vectors failed to stimulate E6-specific T cells despite high level expression of E6/E7 protein. Further experiments revealed that MHC class I presentation of E6/E7 is impaired by the HCMV-vector although it lacks all known immunoevasins. We also generated HCMV-based vectors that express E6-derived peptide fused to HCMV proteins. GBM cells infected with these vectors efficiently stimulated E6-specific T cells. Thus, fusion of antigenic sequences to HCMV proteins is required for efficient presentation via MHC class I molecules during infection. Taken together, these results provide the preclinical basis for development of HCMV-based vaccines and also reveal a novel HCMV-encoded block of MHC class I presentation.
Collapse
Affiliation(s)
- Mohammed O Abdelaziz
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sophia Ossmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Andreas M Kaufmann
- Clinic for Gynecology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, Partner Site Berlin, Berlin, Germany
| | - Martin J Raftery
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Günther Schönrich
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
52
|
Ramadhani AM, Derrick T, Macleod D, Massae P, Malisa A, Mbuya K, Mtuy T, Makupa W, Roberts CH, Bailey RL, Mabey DCW, Holland MJ, Burton MJ. Ocular immune responses, Chlamydia trachomatis infection and clinical signs of trachoma before and after azithromycin mass drug administration in a treatment naïve trachoma-endemic Tanzanian community. PLoS Negl Trop Dis 2019; 13:e0007559. [PMID: 31306419 PMCID: PMC6658141 DOI: 10.1371/journal.pntd.0007559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 07/25/2019] [Accepted: 06/18/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Trachoma, caused by Chlamydia trachomatis, remains the leading infectious cause of blindness worldwide. Persistence and progression of the resulting clinical disease appears to be an immunologically mediated process. Azithromycin, which is distributed at the community level for trachoma control, has immunomodulatory properties. We investigated the impact of one round of oral azithromycin on conjunctival immune responses, C. trachomatis infection and clinical signs three- and six- months post treatment relative to three pre-treatment time-points. METHODOLOGY A cohort of children aged 6 to 10 years were recruited from a trachoma endemic region of northern Tanzania and were visited five times in a 12-month period. They were examined for clinical signs of trachoma and conjunctival swabs were collected for laboratory analysis. C. trachomatis infection was detected and the expression of 46 host genes was quantified using quantitative PCR. All community members were offered azithromycin treatment immediately after the six-month timepoint according to international guidelines. FINDINGS The prevalence of C. trachomatis infection and inflammatory disease signs were significantly reduced three- and six- months post-mass drug administration (MDA). C. trachomatis infection was strongly associated with clinical signs at all five time-points. A profound anti-inflammatory effect on conjunctival gene expression was observed 3 months post-MDA, however, gene expression had largely returned to pre-treatment levels of variation by 6 months. This effect was less marked, but still observed, after adjusting for C. trachomatis infection and when the analysis was restricted to individuals who were free from both infection and clinical disease at all five time-points. Interestingly, a modest effect was also observed in individuals who did not receive treatment. CONCLUSION Conjunctival inflammation is the major clinical risk factor for progressive scarring trachoma, therefore, the reduction in inflammation associated with azithromycin treatment may be beneficial in limiting the development of potentially blinding disease sequelae. Future work should seek to determine whether this effect is mediated directly through inhibition of pro-inflammatory intracellular signalling molecules, through reductions in concurrent, sub-clinical infections, and/or through reduction of infection exposure.
Collapse
Affiliation(s)
- Athumani M. Ramadhani
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Tamsyn Derrick
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - David Macleod
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Aiweda Malisa
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Kelvin Mbuya
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Tara Mtuy
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Chrissy H. Roberts
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Robin L. Bailey
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - David C. W. Mabey
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin J. Holland
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Matthew J. Burton
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
53
|
Osorio C, Kanukuntla T, Diaz E, Jafri N, Cummings M, Sfera A. The Post-amyloid Era in Alzheimer's Disease: Trust Your Gut Feeling. Front Aging Neurosci 2019; 11:143. [PMID: 31297054 PMCID: PMC6608545 DOI: 10.3389/fnagi.2019.00143] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022] Open
Abstract
The amyloid hypothesis, the assumption that beta-amyloid toxicity is the primary cause of neuronal and synaptic loss, has been the mainstream research concept in Alzheimer's disease for the past two decades. Currently, this model is quietly being replaced by a more holistic, “systemic disease” paradigm which, like the aging process, affects multiple body tissues and organs, including the gut microbiota. It is well-established that inflammation is a hallmark of cellular senescence; however, the infection-senescence link has been less explored. Microbiota-induced senescence is a gradually emerging concept promoted by the discovery of pathogens and their products in Alzheimer's disease brains associated with senescent neurons, glia, and endothelial cells. Infectious agents have previously been associated with Alzheimer's disease, but the cause vs. effect issue could not be resolved. A recent study may have settled this debate as it shows that gingipain, a Porphyromonas gingivalis toxin, can be detected not only in Alzheimer's disease but also in the brains of older individuals deceased prior to developing the illness. In this review, we take the position that gut and other microbes from the body periphery reach the brain by triggering intestinal and blood-brain barrier senescence and disruption. We also surmise that novel Alzheimer's disease findings, including neuronal somatic mosaicism, iron dyshomeostasis, aggressive glial phenotypes, and loss of aerobic glycolysis, can be explained by the infection-senescence model. In addition, we discuss potential cellular senescence targets and therapeutic strategies, including iron chelators, inflammasome inhibitors, senolytic antibiotics, mitophagy inducers, and epigenetic metabolic reprograming.
Collapse
Affiliation(s)
- Carolina Osorio
- Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Tulasi Kanukuntla
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Eddie Diaz
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Nyla Jafri
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Michael Cummings
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Adonis Sfera
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| |
Collapse
|
54
|
Assaf S, Hanania NA. Novel therapeutic targets and drug development for the precision treatment of COPD. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1614438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sara Assaf
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nicola A. Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
55
|
Kim NR, Kim YJ. Oxaliplatin regulates myeloid-derived suppressor cell-mediated immunosuppression via downregulation of nuclear factor-κB signaling. Cancer Med 2018; 8:276-288. [PMID: 30592157 PMCID: PMC6346236 DOI: 10.1002/cam4.1878] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022] Open
Abstract
Myeloid‐derived suppressor cells (MDSCs) represent one of the major types of immunoregulatory cells present under abnormal conditions, including cancer. These cells are characterized by their immature phenotype and suppressive effect on various immune effectors. In both human and mouse, there are two main subsets of MDSCs: polymorphonuclear (PMN)‐MDSCs and mononuclear (Mo)‐MDSCs. Thus, strategies to regulate MDSC‐mediated immunosuppression could result in the enhancement of anticancer immune responses. Oxaliplatin, a platinum‐based anticancer agent, is widely used in clinical settings. It is known to induce cell death by interfering with double‐stranded DNA and interrupting its replication and transcription. In this study, we found that oxaliplatin has the potential to regulate MDSC‐mediated immunosuppression in cancer. First, oxaliplatin selectively depleted MDSCs, especially Mo‐MDSCs, but only minimally affected T cells. In addition, sublethal doses of oxaliplatin eliminated the immunosuppressive capacity of MDSCs and induced the differentiation of MDSCs into mature cells. Oxaliplatin treatment diminished the expression of the immunosuppressive functional mediators arginase 1 (ARG1) and NADPH oxidase 2 (NOX2) in MDSCs, while an MDSC‐depleting agent, gemcitabine, did not downregulate these factors significantly. Oxaliplatin‐conditioned MDSCs had no tumor‐promoting activity in vivo. In addition, oxaliplatin modulated the intracellular NF‐κB signaling in MDSCs. Thus, oxaliplatin has the potential to be used as an immunoregulatory agent as well as a cytotoxic drug in cancer treatment.
Collapse
Affiliation(s)
- Na-Rae Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Inje University, Gimhae, Korea
| | - Yeon-Jeong Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Inje University, Gimhae, Korea.,Inje Institute of Pharmaceutical Science and Research, Inje University, Gimhae, Korea
| |
Collapse
|
56
|
Baines KJ, Wright TK, Gibson PG, Powell H, Hansbro PM, Simpson JL. Azithromycin treatment modifies airway and blood gene expression networks in neutrophilic COPD. ERJ Open Res 2018; 4:00031-2018. [PMID: 30406125 PMCID: PMC6215914 DOI: 10.1183/23120541.00031-2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 09/07/2018] [Indexed: 01/06/2023] Open
Abstract
Long-term, low-dose azithromycin reduces exacerbation frequency in chronic obstructive pulmonary disease (COPD), yet the mechanism remains unclear. This study characterised genome-wide gene expression changes in patients with neutrophilic COPD following long-term, low-dose azithromycin treatment. Patients with neutrophilic COPD (>61% or >162×104 cells per mL sputum neutrophils) were randomised to receive either azithromycin or placebo for 12 weeks. Sputum and blood were obtained before and after 12 weeks of treatment. Gene expression was defined using microarrays. Networks were analysed using the Search Tool for the Retrieval of Interacting Gene database. In sputum, 403 genes were differentially expressed following azithromycin treatment (171 downregulated and 232 upregulated), and three following placebo treatment (one downregulated and two upregulated) compared to baseline (adjusted p<0.05 by paired t-test, fold-change >1.5). In blood, 138 genes were differentially expressed with azithromycin (121 downregulated and 17 upregulated), and zero with placebo compared to baseline (adjusted p<0.05 by paired t-test, fold-change >1.3). Network analysis revealed one key network in both sputum (14 genes) and blood (46 genes), involving interferon-stimulated genes, human leukocyte antigens and genes regulating T-cell responses. Long-term, low-dose azithromycin is associated with downregulation of genes regulating antigen presentation, interferon and T-cell responses, and numerous inflammatory pathways in the airways and blood of neutrophilic COPD patients.
Collapse
Affiliation(s)
- Katherine J Baines
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Callaghan, Australia
| | - Thomas K Wright
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Callaghan, Australia
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Callaghan, Australia.,Dept of Respiratory and Sleep Medicine, Hunter New England Area Health Service, Newcastle, Australia
| | - Heather Powell
- Dept of Respiratory and Sleep Medicine, Hunter New England Area Health Service, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Callaghan, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Callaghan, Australia
| |
Collapse
|
57
|
Lagler H, Kiesewetter B, Dolak W, Obermueller M, Simonitsch-Klupp I, Lukas J, Neuper O, Lamm WW, Mayerhoefer ME, Raderer M. Treatment of mucosa associated lymphoid tissue lymphoma with a long-term once-weekly regimen of oral azithromycin: Results from the phase II MALT-A trial. Hematol Oncol 2018; 37:22-26. [PMID: 30153341 PMCID: PMC6585850 DOI: 10.1002/hon.2555] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022]
Abstract
The macrolide clarithromycin has been reported as active for therapy of mucosa associated lymphoid tissue (MALT) lymphoma. Pharmacokinetic properties, however, require continuous daily intake over a prolonged period of time. As the macrolide azithromycin is characterized by a long half‐life as well as potential antineoplastic activity in vitro, we have performed a phase II trial of long‐term once‐weekly oral azithromycin for treatment of MALT lymphoma. In a 2‐stage‐design, 16 patients (10 f/6 m) with histologically verified and measurable MALT lymphoma were included in the first phase of the trial, which could be expanded to a maximum of 46 patients depending on remissions in the first phase. Patients were given oral azithromycin 1500 mg once‐weekly 4 times a month, and restaging was performed after 3 and 6 months. Two patients had gastric and 14 extragastric MALT lymphoma; 12/16 patients were treatment‐naive and received azithromycin as first line treatment. Tolerance of this regimen was excellent, and 14/16 patients received 6 months of treatment as scheduled, while 1 patient each discontinued after 4 (progressive disease) and 1 cycle (personal reasons), respectively. The most commonly observed side effects were mild nausea (n = 8) and diarrhea (n = 4). Efficacy, however, was low as only 4/16 patients (25%) responded, with 2 complete and 2 partial remissions, 9 patients (56%) had stable disease, and 3 patients 19%) were rated as progressive disease. As the predefined activity of more than 7/16 patients responding was not reached, the study was stopped after 16 patients. Although long‐term once‐weekly oral azithromycin showed some antilymphoma activity, the response rate was below the predefined threshold of interest. However, based on our data, one cannot rule out suboptimal dosing in our study; attempts to study azithromycin at a different mode of application might be warranted in the future.
Collapse
Affiliation(s)
- Heimo Lagler
- Department of Medicine 1, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Barbara Kiesewetter
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Werner Dolak
- Department of Medicine III, Clinical Division of Gastroenterology and Hepatology1, Medical University of Vienna, Vienna, Austria
| | - Markus Obermueller
- Department of Medicine 1, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Julius Lukas
- Department of Ophthalmology, Medical University of Vienna, Vienna, Austria
| | - Ortrun Neuper
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang W Lamm
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Marius E Mayerhoefer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Raderer
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
58
|
Fischer M, Bantug GR, Dimeloe S, Gubser PM, Burgener AV, Grählert J, Balmer ML, Develioglu L, Steiner R, Unterstab G, Sauder U, Hoenger G, Hess C. Early effector maturation of naïve human CD8 + T cells requires mitochondrial biogenesis. Eur J Immunol 2018; 48:1632-1643. [PMID: 30028501 DOI: 10.1002/eji.201747443] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 06/08/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022]
Abstract
The role of mitochondrial biogenesis during naïve to effector differentiation of CD8+ T cells remains ill explored. In this study, we describe a critical role for early mitochondrial biogenesis in supporting cytokine production of nascent activated human naïve CD8+ T cells. Specifically, we found that prior to the first round of cell division activated naïve CD8+ T cells rapidly increase mitochondrial mass, mitochondrial respiration, and mitochondrial reactive oxygen species (mROS) generation, which were all inter-linked and important for CD8+ T cell effector maturation. Inhibition of early mitochondrial biogenesis diminished mROS dependent IL-2 production - as well as subsequent IL-2 dependent TNF, IFN-γ, perforin, and granzyme B production. Together, these findings point to the importance of mitochondrial biogenesis during early effector maturation of CD8+ T cells.
Collapse
Affiliation(s)
- Marco Fischer
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Glenn R Bantug
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Sarah Dimeloe
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Patrick M Gubser
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Anne-Valérie Burgener
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Jasmin Grählert
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Maria L Balmer
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Leyla Develioglu
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Rebekah Steiner
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Gunhild Unterstab
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Ursula Sauder
- Microscopy Center, Biocenter, University of Basel, Basel, Switzerland
| | - Gideon Hoenger
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
59
|
Borkner L, Misiak A, Wilk MM, Mills KHG. Azithromycin Clears Bordetella pertussis Infection in Mice but Also Modulates Innate and Adaptive Immune Responses and T Cell Memory. Front Immunol 2018; 9:1764. [PMID: 30105030 PMCID: PMC6077268 DOI: 10.3389/fimmu.2018.01764] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/17/2018] [Indexed: 11/20/2022] Open
Abstract
Treatment with the macrolide antibiotic azithromycin (AZM) is an important intervention for controlling infection of children with Bordetella pertussis and as a prophylaxis for preventing transmission to family members. However, antibiotics are known to have immunomodulatory effects independent of their antimicrobial activity. Here, we used a mouse model to examine the effects of AZM treatment on clearance of B. pertussis and induction of innate and adaptive immunity. We found that treatment of mice with AZM either 7 or 14 days post challenge effectively cleared the bacteria from the lungs. The numbers of innate immune cells in the lungs were significantly reduced in antibiotic-treated mice. Furthermore, AZM reduced the activation status of macrophages and dendritic cells, but only in mice treated on day 7. Early treatment with antibiotics also reduced the frequency of tissue-resident T cells and IL-17-producing cells in the lungs. To assess the immunomodulatory effects of AZM independent of its antimicrobial activity, mice were antibiotic treated during immunization with a whole cell pertussis (wP) vaccine. Protection against B. pertussis induced by immunization with wP was slightly reduced in AZM-treated mice. Antibiotic-treated wP-immunized mice had reduced numbers of lung-resident memory CD4 T cells and IL-17-production and reduced CD49d expression on splenic CD4 T cells after challenge, suggestive of impaired CD4 T cell memory. Taken together these results suggest that AZM can modulate the induction of memory CD4 T cells during B. pertussis infection, but this may in part be due to the clearance of B. pertussis and resulting loss of components that stimulate innate and adaptive immune response.
Collapse
Affiliation(s)
- Lisa Borkner
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alicja Misiak
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mieszko M Wilk
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
60
|
Effects of pre-transplant azithromycin administration on kidney graft function: study protocol for a double-blind randomized clinical trial. Trials 2018; 19:345. [PMID: 29950182 PMCID: PMC6022710 DOI: 10.1186/s13063-018-2744-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/14/2018] [Indexed: 12/02/2022] Open
Abstract
Background Kidney transplantation is the best strategy for the management of end-stage renal disease; however, the outcomes need to improve further. Macrolides show antimicrobial and anti-inflammatory properties in chronic diseases and intraoperatively, and can accumulate in tissues for extended periods. Therefore, theoretically, when administered to a donor and because of accumulation in the donor kidney, macrolides can cause graft immunomodulation and improve kidney transplantation outcomes. Methods This study is a single-center, randomized clinical trial. A total of 60 kidney donors will be randomly allocated to the azithromycin or placebo group and treated with a single dose (1 g) of azithromycin or placebo, respectively, 1 day before surgery. Recruitment commenced in September 2016 and is expected to be completed by March 2018. The primary outcome is kidney graft function. The secondary outcomes include rejection rate, urinary tract infections in graft recipients, pain and systemic inflammatory response syndrome in live donors, and complications in both donors and recipients. Outcomes will be evaluated at baseline and every day in the first week after transplantation, as well as at 1 and 3 months post transplantation. Adverse reactions will be documented. If the efficacy of azithromycin in reducing adverse outcomes is confirmed, it would serve as an easy to use, economic intervention able to lower post-transplantation risks. Discussion Short and mid-term analyses of blood and urine samples as well as immunological assays will facilitate a more in-depth analysis of the effects of azithromycin on transplantation outcomes. Trial registration Iranian Clinical Trial Registry, IRCT201606141853N11, registered on September 5, 2016. Electronic supplementary material The online version of this article (10.1186/s13063-018-2744-y) contains supplementary material, which is available to authorized users.
Collapse
|
61
|
|
62
|
First-line antibiotic therapy in Helicobacter pylori-negative low-grade gastric mucosa-associated lymphoid tissue lymphoma. Sci Rep 2017; 7:14333. [PMID: 29084984 PMCID: PMC5662601 DOI: 10.1038/s41598-017-14102-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 10/02/2017] [Indexed: 12/13/2022] Open
Abstract
First-line antibiotic treatment for eradicating Helicobacter pylori (HP) infection is effective in HP-positive low-grade gastric mucosa-associated lymphoid tissue lymphoma (MALToma), but its role in HP-negative cases is uncertain. In this exploratory retrospective study, we assessed the outcome and potential predictive biomarkers for 25 patients with HP-negative localized gastric MALToma who received first-line HP eradication (HPE) therapy. An HP-negative status was defined as negative results on histology, rapid urease test, 13C urea breath test, and serology. We observed an antibiotic response (complete remission [CR], number = 8; partial remission, number = 1) in 9 (36.0%) out of 25 patients. A t(11;18)(q21;q21) translocation was detected in 7 (43.8%) of 16 antibiotic-unresponsive cases, but in none of the 9 antibiotic-responsive cases (P = 0.027). Nuclear BCL10 expression was significantly higher in antibiotic-unresponsive tumors than in antibiotic-responsive tumors (14/16 [87.5%] vs. 1/9 [11.1%]; P = 0.001). Nuclear NF-κB expression was also significantly higher in antibiotic-unresponsive tumors than in antibiotic-responsive tumors (12/16 [75.0%] vs. 1/9 [11.1%]; P = 0.004). A substantial portion of patients with HP-negative gastric MALToma responded to first-line HPE. In addition to t(11;18)(q21;q21), BCL10 and NF-κB are useful immunohistochemical biomarkers to predict antibiotic-unresponsive status in this group of tumors.
Collapse
|
63
|
Ruiz VE, Battaglia T, Kurtz ZD, Bijnens L, Ou A, Engstrand I, Zheng X, Iizumi T, Mullins BJ, Müller CL, Cadwell K, Bonneau R, Perez-Perez GI, Blaser MJ. A single early-in-life macrolide course has lasting effects on murine microbial network topology and immunity. Nat Commun 2017; 8:518. [PMID: 28894149 PMCID: PMC5593929 DOI: 10.1038/s41467-017-00531-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022] Open
Abstract
Broad-spectrum antibiotics are frequently prescribed to children. Early childhood represents a dynamic period for the intestinal microbial ecosystem, which is readily shaped by environmental cues; antibiotic-induced disruption of this sensitive community may have long-lasting host consequences. Here we demonstrate that a single pulsed macrolide antibiotic treatment (PAT) course early in life is sufficient to lead to durable alterations to the murine intestinal microbiota, ileal gene expression, specific intestinal T-cell populations, and secretory IgA expression. A PAT-perturbed microbial community is necessary for host effects and sufficient to transfer delayed secretory IgA expression. Additionally, early-life antibiotic exposure has lasting and transferable effects on microbial community network topology. Our results indicate that a single early-life macrolide course can alter the microbiota and modulate host immune phenotypes that persist long after exposure has ceased.High or multiple doses of macrolide antibiotics, when given early in life, can perturb the metabolic and immunological development of lab mice. Here, Ruiz et al. show that even a single macrolide course, given early in life, leads to long-lasting changes in the gut microbiota and immune system of mice.
Collapse
Affiliation(s)
- Victoria E Ruiz
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Thomas Battaglia
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Zachary D Kurtz
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Luc Bijnens
- Janssen R&D, Janssen Pharmaceutical Companies of J&J, Turnhoutseweg 30, Beerse, 2340, Belgium
| | - Amy Ou
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Isak Engstrand
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Xuhui Zheng
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Tadasu Iizumi
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Briana J Mullins
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Christian L Müller
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, 10010, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, NYUSM, New York, NY, 10016, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, 10010, USA.,Department of Biology, Center for Genomics and Systems Biology, NYU, New York, NY, 10003, USA.,Courant Institute of Mathematical Sciences, NYU, New York, NY, 10012, USA
| | - Guillermo I Perez-Perez
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Martin J Blaser
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA. .,New York Harbor Department of Veterans Affairs Medical Center, New York, NY, 10010, USA.
| |
Collapse
|
64
|
Gibson PG, Yang IA, Upham JW, Reynolds PN, Hodge S, James AL, Jenkins C, Peters MJ, Marks GB, Baraket M, Powell H, Taylor SL, Leong LEX, Rogers GB, Simpson JL. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): a randomised, double-blind, placebo-controlled trial. Lancet 2017; 390:659-668. [PMID: 28687413 DOI: 10.1016/s0140-6736(17)31281-3] [Citation(s) in RCA: 427] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Exacerbations of asthma cause a substantial global illness burden. Adults with uncontrolled persistent asthma despite maintenance treatment require additional therapy. Since macrolide antibiotics can be used to treat persistent asthma, we aimed to assess the efficacy and safety of oral azithromycin as add-on therapy in patients with uncontrolled persistent asthma on medium-to-high dose inhaled corticosteroids plus a long-acting bronchodilator. METHODS We did a randomised, double-blind, placebo controlled parallel group trial to determine whether oral azithromycin decreases the frequency of asthma exacerbations in adults (≥18 years) with symptomatic asthma despite current use of inhaled corticosteroid and long-acting bronchodilator, and who had no hearing impairment or abnormal prolongation of the corrected QT interval. Patients were randomly assigned (1:1) to receive azithromycin 500 mg or placebo three times per week for 48 weeks. Patients were centrally allocated using concealed random allocation from a computer-generated random numbers table with permuted blocks of 4 or 6 and stratification for centre and past smoking. Primary efficacy endpoints were the rate of total (severe and moderate) asthma exacerbations over 48 weeks and asthma quality of life. Data were analysed on an intention-to-treat basis. The trial is registered at the Australian and New Zealand Clinical Trials Registry (ANZCTR), number 12609000197235. FINDINGS Between June 12, 2009, and Jan 31, 2015, 420 patients were randomly assigned (213 in the azithromycin group and 207 in the placebo group). Azithromycin reduced asthma exacerbations (1·07 per patient-year [95% CI 0·85-1·29]) compared with placebo (1·86 per patient-year [1·54-2·18]; incidence rate ratio [IRR] 0·59 [95% CI 0·47-0·74]; p<0·0001). The proportion of patients experiencing at least one asthma exacerbation was reduced by azithromycin treatment (127 [61%] patients in the placebo group vs 94 [44%] patients in the azithromycin group, p<0·0001). Azithromycin significantly improved asthma-related quality of life (adjusted mean difference, 0·36 [95% CI 0·21-0·52]; p=0·001). Diarrhoea was more common in azithromycin-treated patients (72 [34%] vs 39 [19%]; p=0·001). INTERPRETATION Adults with persistent symptomatic asthma experience fewer asthma exacerbations and improved quality of life when treated with oral azithromycin for 48 weeks. Azithromycin might be a useful add-on therapy in persistent asthma. FUNDING National Health and Medical Research Council of Australia, John Hunter Hospital Charitable Trust.
Collapse
Affiliation(s)
- Peter G Gibson
- Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, Newcastle, NSW, Australia; Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; Woolcock Institute of Medical Research, Sydney, NSW, Australia.
| | - Ian A Yang
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - John W Upham
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Paul N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia; Lung Research Laboratory, Hanson Institute, Adelaide, SA, Australia; School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Sandra Hodge
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia; Lung Research Laboratory, Hanson Institute, Adelaide, SA, Australia; School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia; School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia
| | - Christine Jenkins
- Respiratory Trials, The George Institute for Global Health, Sydney, NSW, Australia; Department of Thoracic Medicine, Concord General Hospital, Sydney, NSW, Australia
| | - Matthew J Peters
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia; Department of Thoracic Medicine, Concord General Hospital, Sydney, NSW, Australia
| | - Guy B Marks
- Woolcock Institute of Medical Research, Sydney, NSW, Australia; South Western Sydney Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Melissa Baraket
- Respiratory Medicine Department and Ingham Institute Liverpool Hospital, University of New South Wales Medicine Faculty, Sydney, NSW, Australia
| | - Heather Powell
- Department of Respiratory and Sleep Medicine, Hunter Medical Research Institute, Newcastle, NSW, Australia; Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Steven L Taylor
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia; SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Lex E X Leong
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia; SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Geraint B Rogers
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia; SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
65
|
Kim J, Lee KP, Lee DW, Lim K. Piperine enhances carbohydrate/fat metabolism in skeletal muscle during acute exercise in mice. Nutr Metab (Lond) 2017; 14:43. [PMID: 28680454 PMCID: PMC5496355 DOI: 10.1186/s12986-017-0194-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/07/2017] [Indexed: 12/31/2022] Open
Abstract
Background Exercise promotes energy metabolism (e.g., metabolism of glucose and lipids) in skeletal muscles; however, reactive oxygen species are also generated during exercise. Various spices have been reported to have beneficial effects in sports medicine. Here, we investigated the effects of piperine, an active compound in black pepper, to determine its effects on metabolism during acute endurance exercise. Methods ICR mice (n = 18) were divided into three groups: nonexercise (CON), exercise (EX), and exercise with piperine (5 mg/kg) treatment (EP). Mice were subjected to enforced exercise on a treadmill at a speed of 22 m/min for 1 h. To evaluate the inflammatory responses following exercise, fluorescence-activated cell sorting analysis was performed to monitor changes in CD4+ cells within the peripheral blood mononuclear cells (PBMCs) of mice. The expression levels of metabolic pathway components and redox-related factors were evaluated in the soleus muscle by reverse transcription polymerase chain reaction and western blotting. Results There were no changes in the differentiation of immune cells in PBMCs in both the EX and EP groups compared with that in the CON group. Mice in the EX group exhibited a significant increase in the expression of metabolic pathway components and redox signal-related components compared with mice in the CON group. Moreover, mice in the EP group showed greater metabolic (GLUT4, MCT1, FAT/CD36, CPT1, CS) changes than mice in the EX group, and changes in the expression of redox signal components were lower in the EP group than those in the EX group. Conclusion Our findings demonstrate that piperine promoted beneficial metabolism during exercise by regulating carbohydrate/fat metabolism and redox signals. Therefore, piperine may be a candidate supplement for improvement of exercise ability. Electronic supplementary material The online version of this article (doi:10.1186/s12986-017-0194-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jisu Kim
- Physical Activity & Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Kang-Pa Lee
- Department of Medical Science, School of Medicine Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Dae-Won Lee
- Department of Bio-Science, College of Natural Science, Dongguk University, Dongdae-ro 123, Gyeongju, Gyeongsangbuk-do 38066 Republic of Korea
| | - Kiwon Lim
- Physical Activity & Performance Institute, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea.,Department of Physical Education, Laboratory of Exercise Nutrition, Korea University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 143-701 Republic of Korea
| |
Collapse
|
66
|
Battin C, Hennig A, Mayrhofer P, Kunert R, Zlabinger GJ, Steinberger P, Paster W. A human monocytic NF-κB fluorescent reporter cell line for detection of microbial contaminants in biological samples. PLoS One 2017; 12:e0178220. [PMID: 28542462 PMCID: PMC5443541 DOI: 10.1371/journal.pone.0178220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/09/2017] [Indexed: 12/16/2022] Open
Abstract
Sensing of pathogens by innate immune cells is essential for the initiation of appropriate immune responses. Toll-like receptors (TLRs), which are highly sensitive for various structurally and evolutionary conserved molecules derived from microbes have a prominent role in this process. TLR engagement results in the activation of the transcription factor NF-κB, which induces the expression of cytokines and other inflammatory mediators. The exquisite sensitivity of TLR signalling can be exploited for the detection of bacteria and microbial contaminants in tissue cultures and in protein preparations. Here we describe a cellular reporter system for the detection of TLR ligands in biological samples. The well-characterized human monocytic THP-1 cell line was chosen as host for an NF-ᴋB-inducible enhanced green fluorescent protein reporter gene. We studied the sensitivity of the resultant reporter cells for a variety of microbial components and observed a strong reactivity towards TLR1/2 and TLR2/6 ligands. Mycoplasma lipoproteins are potent TLR2/6 agonists and we demonstrate that our reporter cells can be used as reliable and robust detection system for mycoplasma contaminations in cell cultures. In addition, a TLR4-sensitive subline of our reporters was engineered, and probed with recombinant proteins expressed in different host systems. Bacterially expressed but not mammalian expressed proteins induced strong reporter activity. We also tested proteins expressed in an E. coli strain engineered to lack TLR4 agonists. Such preparations also induced reporter activation in THP-1 cells highlighting the importance of testing recombinant protein preparations for microbial contaminations beyond endotoxins. Our results demonstrate the usefulness of monocytic reporter cells for high-throughput screening for microbial contaminations in diverse biological samples, including tissue culture supernatants and recombinant protein preparations. Fluorescent reporter assays can be measured on standard flow cytometers and in contrast to established detection methods, like luciferase-based systems or Limulus Amebocyte Lysate tests, they do not require costly reagents.
Collapse
Affiliation(s)
- Claire Battin
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika Hennig
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Patrick Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gerhard J. Zlabinger
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail: (PS); (WP), (WP)
| | - Wolfgang Paster
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- * E-mail: (PS); (WP), (WP)
| |
Collapse
|
67
|
Lee N, Wong CK, Chan MCW, Yeung ESL, Tam WWS, Tsang OTY, Choi KW, Chan PKS, Kwok A, Lui GCY, Leung WS, Yung IMH, Wong RYK, Cheung CSK, Hui DSC. Anti-inflammatory effects of adjunctive macrolide treatment in adults hospitalized with influenza: A randomized controlled trial. Antiviral Res 2017; 144:48-56. [PMID: 28535933 DOI: 10.1016/j.antiviral.2017.05.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022]
Abstract
INTRODUCTION - Macrolides can ameliorate inflammation in respiratory diseases, providing clinical benefits. Data in influenza is lacking. METHOD - A randomized, open-label, multicenter trial among adults hospitalized for laboratory-confirmed influenza was conducted. Study treatments of oseltamivir and azithromycin (500 mg/day), or oseltamivir alone, both for 5 days, were allocated at 1:1 ratio. The primary outcome was plasma cytokine/chemokine concentration change over time (Day 0-10); secondary outcomes were viral load and symptom score changes. Generalized Estimating Equation (GEE) models were used to analyze longitudinal data. RESULTS - Fifty patients were randomized to the oseltamivir-azithromycin or oseltamivir groups, with comparable baseline characteristics (age, 57 ± 18 years; A/H3N2, 70%), complications (72%), and viral load. Pro-inflammatory cytokines IL-6 (GEE: β -0.037, 95%CI-0.067,-0.007, P = 0.016; reduction from baseline -83.4% vs -59.5%), CXCL8/IL-8 (β -0.018, 95%CI-0.037,0.000, P = 0.056; -80.5% vs -58.0%), IL-17 (β -0.064, 95%CI-0.117,-0.012, P = 0.015; -74.0% vs -34.3%), CXCL9/MIG (β -0.010, 95%CI-0.020,0.000, P = 0.043; -71.3% vs -56.0%), sTNFR-1, IL-18, and CRP declined faster in the oseltamivir-azithromycin group. There was a trend toward faster symptom resolution (β -0.463, 95%CI-1.297,0.371). Viral RNA decline (P = 0.777) and culture-negativity rates were unaffected. Additional ex vivo studies confirmed reduced induction of IL-6 (P = 0.017) and CXCL8/IL-8 (P = 0.005) with azithromycin. CONCLUSION - We found significant anti-inflammatory effects with adjunctive macrolide treatment in adults with severe influenza infections. Virus control was unimpaired. Clinical benefits of a macrolide-containing regimen deserve further study. [ClinicalTrials.gov NCT01779570].
Collapse
Affiliation(s)
- Nelson Lee
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong; Stanley Ho Center for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong.
| | - Chun-Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong
| | - Martin C W Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong
| | - Esther S L Yeung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong; Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong
| | - Wilson W S Tam
- Alice Lee Centre for Nursing Studies, National University of Singapore, Singapore
| | - Owen T Y Tsang
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Kin-Wing Choi
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Hong Kong
| | - Paul K S Chan
- Stanley Ho Center for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong; Department of Microbiology, The Chinese University of Hong Kong, Hong Kong
| | - Angela Kwok
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong
| | - Grace C Y Lui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Wai-Shing Leung
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Irene M H Yung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Rity Y K Wong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong; Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Catherine S K Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - David S C Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong; Stanley Ho Center for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
68
|
Jutz S, Hennig A, Paster W, Asrak Ö, Dijanovic D, Kellner F, Pickl WF, Huppa JB, Leitner J, Steinberger P. A cellular platform for the evaluation of immune checkpoint molecules. Oncotarget 2017; 8:64892-64906. [PMID: 29029399 PMCID: PMC5630299 DOI: 10.18632/oncotarget.17615] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Blockade of the T cell coinhibitory molecules CTLA-4 and PD-1 has clinical utility to strengthen T cell responses. In addition to these immune checkpoints an ever-growing number of molecules has been implicated in generating coinhibitory signals in T cells. However, investigating coinhibitory molecules in primary human cells is complicated by the restricted expression and promiscuity of both coinhibitory receptors and their ligands. Here we have evaluated the potential of fluorescence-based transcriptional reporters based on the human Jurkat T cell line in conjunction with engineered T cell stimulator cell lines for investigating coinhibitory pathways. CTLA-4, PD-1, TIGIT, BTLA and 2B4 expressing reporter cells were generated and activated with T cell stimulator cells expressing cognate ligands of these molecules. All accessory molecules tested were functional in our reporter system. Engagement of CTLA-4, PD-1, BTLA and TIGIT by their ligands significantly inhibited T cell activation, whereas binding of 2B4 by CD48 resulted in enhanced responses. Mutational analysis revealed intracellular motifs that are responsible for BTLA mediated T cell inhibition and demonstrates potent reporter inhibition by CTLA-4 independent of cytoplasmic signaling motifs. Moreover, considerably higher IC50 values were measured for the CTLA-4 blocker Ipilimumab compared to the PD-1 antibody Nivolumab. Our findings show that coinhibitory pathways can be evaluated in Jurkat-based transcriptional reporters and yield novel insights on their function. Results obtained from this robust reductionist system can complement more time consuming and complex studies of such pathways in primary T cells.
Collapse
Affiliation(s)
- Sabrina Jutz
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika Hennig
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Paster
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ömer Asrak
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dejana Dijanovic
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Florian Kellner
- Department of Molecular Immunology, Immune Recognition Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Division of Cellular Immunology and Immunohematology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes B Huppa
- Department of Molecular Immunology, Immune Recognition Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
69
|
Tauber PA, Pickl WF. Pharmacological targeting of allergen-specific T lymphocytes. Immunol Lett 2017; 189:27-39. [PMID: 28322861 DOI: 10.1016/j.imlet.2017.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022]
Abstract
Allergic disorders are the result of a complex pathophysiology, involving major cellular lineages and a multitude of humoral factors of the innate and adaptive immune system, and have the tendency to involve multiple organs. Consequently, even standard pharmacological treatment of allergies is rarely specific but usually targets more than one pathway/cellular system at a time. Accordingly, many of the classic anti-allergic drugs have a critical impact also on T helper cells, which are pivotal not only during the sensitization but also the maintenance phase of allergic diseases. Recent years have seen a dramatic increase of novel drugs with the potency to interfere, more or less specifically, with T lymphocyte function, which might, possibly together with classic anti-allergic drugs, help harnessing one of the central cellular players in allergic responses. A major theme in the years to come will be a thoughtful combination of previously established with recently developed treatment modalities.
Collapse
Affiliation(s)
- Peter A Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
70
|
Chloroquine inhibits human CD4 + T-cell activation by AP-1 signaling modulation. Sci Rep 2017; 7:42191. [PMID: 28169350 PMCID: PMC5294581 DOI: 10.1038/srep42191] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/06/2017] [Indexed: 12/17/2022] Open
Abstract
Chloroquine (CQ) is widely used as an anti-inflammatory therapeutic for rheumatic diseases. Although its modes of action on the innate immune system are well described, there is still insufficient knowledge about its direct effects on the adaptive immune system. Thus, we evaluated the influence of CQ on activation parameters of human CD4+ T-cells. CQ directly suppressed proliferation, metabolic activity and cytokine secretion of T-cells following anti-CD3/anti-CD28 activation. In contrast, CQ showed no effect on up-regulation of T-cell activation markers. CQ inhibited activation of all T helper cell subsets, although IL-4 and IL-13 secretion by Th2 cells were less influenced compared to other Th-specific cytokines. Up to 10 μM, CQ did not reduce cell viability, suggesting specific suppressive effects on T-cells. These properties of CQ were fully reversible in re-stimulation experiments. Analyses of intracellular signaling showed that CQ specifically inhibited autophagic flux and additionally activation of AP-1 by reducing phosphorylation of c-JUN. This effect was mediated by inhibition of JNK catalytic activity. In summary, we characterized selective and reversible immunomodulatory effects of CQ on human CD4+ T-cells. These findings provide new insights into the biological actions of JNK/AP-1 signaling in T-cells and may help to expand the therapeutic spectrum of CQ.
Collapse
|
71
|
Awan S, Crosby V, Potter V, Hennig I, Baldwin D, Ndlovu M, Paradine S, Wilcock A. Is clarithromycin a potential treatment for cachexia in people with lung cancer? A feasibility study. Lung Cancer 2016; 104:75-78. [PMID: 28213005 DOI: 10.1016/j.lungcan.2016.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/16/2016] [Accepted: 12/18/2016] [Indexed: 11/25/2022]
Abstract
Clarithromycin may improve cachexia and survival in non-small cell lung cancer (NSCLC), but adequately controlled data are lacking. This study was undertaken primarily to inform the feasibility and scale of a phase III trial. Eligible consenting patients with stage IV NSCLC and cachexia were to be randomized to receive either clarithromycin 250mg twice daily or placebo for eight weeks. Aspects of trial feasibility recorded included numbers eligible, approached and recruited, together with adherence and completion of treatment and assessments. Over 6 months, none of 125 patients identified fulfilled the entry criteria. The commonest reasons for ineligibility were the use of an excluded concurrent drug (45, 36%), brain metastases (22, 18%), poor performance status (21, 17%) and current chemotherapy (15, 12%). A phase III trial of clarithromycin using these entry criteria is not feasible in this setting. Other macrolides that have a lower risk of a drug-drug interaction may be more practical to pursue.
Collapse
Affiliation(s)
- Sarah Awan
- Department of Palliative Care, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Vincent Crosby
- Department of Palliative Care, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Vanessa Potter
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Ivo Hennig
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - David Baldwin
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Mehluli Ndlovu
- Nottingham Health Science Partners, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Sharon Paradine
- Consumer representative, c/o National Cancer Research Institute, Angel Building, 407, St. John Street, London, EC1V 4AD, UK
| | - Andrew Wilcock
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK.
| |
Collapse
|
72
|
Czyżewska-Dors E, Kwit K, Pejsak Z, Pomorska-Mól M. Changes in circulating lymphocyte subpopulations in pigs receiving therapeutic doses of ceftiofur and tulathromycin. J Vet Res 2016. [DOI: 10.1515/jvetres-2016-0071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Introduction: The aim of the study was to evaluate the effect of administration of therapeutic doses of ceftiofur and tulathromycin on the circulating lymphocyte subpopulations in healthy pigs. Material and Methods: The study was conducted on thirty healthy 7- to 10-week-old pigs, assigned to three groups: the TUL group, injected with tulathromycin (n = 10); the CEF group, injected with ceftiofur (n = 10); and the C group, the control with no antibiotic administration (n = 10). Blood samples were collected before, during, and after treatment with antimicrobials. Lymphocyte subpopulations circulating in the blood were determined by immunostaining and flow cytometry analyses. Results: Following administration of a therapeutic dose of tulathromycin, there were no changes in the lymphocyte subpopulations circulating in blood. In contrast, administration of ceftiofur at the recommended dose decreased the absolute number of CD3+, CD21+, CD4+CD8-, CD4-CD8+, and double positive CD4CD8 cells. Conclusion: Results from the study indicate that ceftiofur possesses the ability to modulate the immune system in healthy pigs by decreasing lymphocyte subpopulations circulating in blood.
Collapse
Affiliation(s)
- Ewelina Czyżewska-Dors
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Krzysztof Kwit
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Zygmunt Pejsak
- Department of Swine Diseases, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | | |
Collapse
|
73
|
Lin SJ, Kuo ML, Hsiao HS, Lee PT. Azithromycin modulates immune response of human monocyte-derived dendritic cells and CD4 + T cells. Int Immunopharmacol 2016; 40:318-326. [PMID: 27664570 DOI: 10.1016/j.intimp.2016.09.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/29/2016] [Accepted: 09/14/2016] [Indexed: 02/03/2023]
Abstract
Azithromycin (AZM) is a macrolide antibiotic that exhibits anti-inflammatory activity aside from its antimicrobial effect, a feature that may ameliorate certain inflammatory disorders and prevent graft-versus-host disease in patients receiving stem cell transplantation. In the present study, we investigated the ability of AZM to influence the function of human monocyte-derived dendritic cells (DCs) and CD4+ T cells. We found that AZM down-regulated CD80, CD86, and HLA-DR expression in lipopolysaccharide (LPS)-stimulated DCs and suppressed interleukin (IL)-6, IL-10, IL-12, and tumor necrosis factor-alpha production in these cells. In addition, AZM increased endocytosis and/or expression of Toll-like receptor (TLR)2, TLR4, and TLR9 in DCs and suppressed anti-CD3/CD28-induced CD4+ T cell proliferation and interferon-gamma production, an effect that was synergistic with dexamethasone. Finally, AZM suppressed DC-induced allogeneic T cell proliferation and cytokine production. Our study demonstrates that AZM modulates DC and CD4+ T cell function and may be of therapeutic benefit in various inflammatory disorders.
Collapse
Affiliation(s)
- Syh-Jae Lin
- Division of Asthma, Allergy, and Rheumatology, Department of Pediatrics, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan; Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Hsiu-Shan Hsiao
- Division of Asthma, Allergy, and Rheumatology, Department of Pediatrics, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Pei-Tzu Lee
- Division of Asthma, Allergy, and Rheumatology, Department of Pediatrics, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
74
|
Modak M, Majdic O, Cejka P, Jutz S, Puck A, Gerwien JG, Steinberger P, Zlabinger GJ, Strobl H, Stöckl J. Engagement of distinct epitopes on CD43 induces different co-stimulatory pathways in human T cells. Immunology 2016; 149:280-296. [PMID: 27392084 PMCID: PMC5046061 DOI: 10.1111/imm.12642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 06/07/2016] [Accepted: 06/24/2016] [Indexed: 12/30/2022] Open
Abstract
Co‐receptors, being either co‐stimulatory or co‐inhibitory, play a pivotal role in T‐cell immunity. Several studies have indicated that CD43, one of the abundant T‐cell surface glycoproteins, acts not only as a potent co‐receptor but also as a negative regulator for T‐cell activation. Here we demonstrate that co‐stimulation of human peripheral blood (PB) T cells through two distinct CD43 epitopes recognized by monoclonal antibodies (mAb) CD43‐6E5 (T6E5‐act) and CD43‐10G7 (T10G7‐act) potently induced T‐cell proliferation. However, T‐cell co‐stimulation through two CD43 epitopes differentially regulated activation of nuclear factor of activated T cells (NFAT) and nuclear factor‐κB (NF‐κB) transcription factors, T‐cell cytokine production and effector function. T6E5‐act produced high levels of interleukin‐22 (IL‐22) and interferon‐γ (IFN‐γ) similar to T cells activated via CD28 (TCD28‐act), whereas T10G7‐act produced low levels of inflammatory cytokines but higher levels of regulatory cytokines transforming growth factor‐β (TGF‐β) and interleukin‐35 (IL‐35). Compared with T6E5‐act or to TCD28‐act, T10G7‐act performed poorly in response to re‐stimulation and further acquired a T‐cell suppressive function. T10G7‐act did not directly inhibit proliferation of responder T cells, but formed stable heterotypic clusters with dendritic cells (DC) via CD2 to constrain activation of responder T cells. Together, our data demonstrate that CD43 is a unique and polarizing regulator of T‐cell function.
Collapse
Affiliation(s)
- Madhura Modak
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Otto Majdic
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Petra Cejka
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabrina Jutz
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alexander Puck
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jens G Gerwien
- Biopharmaceuticals Research Unit, Inflammation Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Peter Steinberger
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Institute of Pathophysiology and Immunology, Centre of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Johannes Stöckl
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
75
|
Ohe M, Hashino S. Successful treatment of angioimmunoblastic T-cell lymphoma with clarithromycin. Blood Res 2016; 51:139-42. [PMID: 27382561 PMCID: PMC4931934 DOI: 10.5045/br.2016.51.2.139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 07/25/2015] [Indexed: 11/17/2022] Open
Affiliation(s)
- Masashi Ohe
- Department of General Medicine, JCHO Hokkaido Hospital, Sapporo, Japan
| | | |
Collapse
|
76
|
Jutz S, Leitner J, Schmetterer K, Doel-Perez I, Majdic O, Grabmeier-Pfistershammer K, Paster W, Huppa JB, Steinberger P. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: Simultaneous measurement of NF-κB, NFAT and AP-1. J Immunol Methods 2016; 430:10-20. [PMID: 26780292 DOI: 10.1016/j.jim.2016.01.007] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 12/21/2015] [Accepted: 01/13/2016] [Indexed: 12/29/2022]
Abstract
Engagement of the T cell receptor complex reprograms T cells for proliferation, cytokine production and differentiation towards effector cells. This process depends on activating costimulatory signals and is counteracted by coinhibitory molecules. Three transcription factors, namely NF-κB, NFAT and AP-1, have a major role in inducing the transcriptional program that is required for T cell activation and differentiation. Here we describe the generation of a triple parameter reporter based on the human Jurkat T cell line, where response elements for NF-κB, NFAT and AP-1 drive the expression of the fluorescent proteins CFP, eGFP and mCherry, respectively. The emission spectra of these proteins allow simultaneous assessment of NF-κB, NFAT and AP-1 activity in response to stimulation. Ligation of the TCR complex induced moderate reporter activity, which was strongly enhanced upon coengagement of the costimulatory receptors CD2 or CD28. Moreover, we have generated and tested triple parameter reporter cells that harbor costimulatory and inhibitory receptors not endogenously expressed in the Jurkat cells. In these experiments we could show that engagement of the costimulatory molecule 4-1BB enhances NF-κB and AP-1 activity, whereas coinhibition via PD-1 or BTLA strongly reduced the activation of NF-κB and NFAT. Engagement of BTLA significantly inhibited AP-1, whereas PD-1 had little effect on the activation of this transcription factor. Our triple parameter reporter T cell line is an excellent tool to assess the effect of costimulatory and coinhibitory receptors on NF-κB, NFAT and AP-1 activity and has a wide range of applications beyond the evaluation of costimulatory pathways.
Collapse
Affiliation(s)
- Sabrina Jutz
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Klaus Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Iago Doel-Perez
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Otto Majdic
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Wolfgang Paster
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes B Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
77
|
Prodinger J, Loacker LJ, Schmidt RLJ, Ratzinger F, Greiner G, Witzeneder N, Hoermann G, Jutz S, Pickl WF, Steinberger P, Marculescu R, Schmetterer KG. The tryptophan metabolite picolinic acid suppresses proliferation and metabolic activity of CD4+ T cells and inhibits c-Myc activation. J Leukoc Biol 2015; 99:583-94. [PMID: 26497245 DOI: 10.1189/jlb.3a0315-135r] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 10/05/2015] [Indexed: 11/24/2022] Open
Abstract
Tryptophan metabolites, including kynurenine, 3-hydroxyanthranilic acid, and picolinic acid, are key mediators of immunosuppression by cells expressing the tryptophan-catabolizing enzyme indoleamine2,3-dioxygenase. In this study, we assessed the influence of picolinic acid on cell viability and effector functions of CD4(+)T cells following in vitro activation with agonistic anti-CD3/anti-CD28 antibodies. In contrast to kynurenine and 3-hydroxyanthranilic acid, exposure of T cells with picolinic acid did not affect cell viability, whereas proliferation and metabolic activity were suppressed in a dose-dependent manner. On the other hand, cytokine secretion and up-regulation of cell surface activation markers were not or only weakly inhibited by picolinic acid. Picolinic acid exposure induced a state of deep anergy that could not be overcome by the addition of exogenous IL-2 and inhibited Th cell polarization. On the molecular level, important upstream signaling molecules, such as the MAPKs ERK and p38 and the mammalian target of rapamycin target protein S6 ribosomal protein, were not affected by picolinic acid. Likewise, NFAT, NF-κB, and AP-1 promoter activity in Jurkat T cells was not influenced by exposure to picolinic acid. Whereas transcriptional levels of v-myc avian myelocytomatosis viral oncogene homolog were not affected by picolinic acid, phosphorylation at Ser62 was strongly reduced in picolinic acid-exposed T cells following activation. In conclusion, picolinic acid mediates a unique immunosuppressive program in T cells, mainly inhibiting cell cycle and metabolic activity, while leaving other effector functions intact. These functional features are accompanied by reduced phosphorylation of v-myc avian myelocytomatosis viral oncogene homolog. It remains to be determined whether this effect is mediated by direct inhibition of ERK activity or whether indirect mechanisms apply.
Collapse
Affiliation(s)
- Johanna Prodinger
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Lisa J Loacker
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Ralf L J Schmidt
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Franz Ratzinger
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Georg Greiner
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Nadine Witzeneder
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabrina Jutz
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Rodrig Marculescu
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Klaus G Schmetterer
- *Department of Laboratory Medicine and Institute of Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
78
|
Gualdoni GA, Lingscheid T, Schmetterer KG, Hennig A, Steinberger P, Zlabinger GJ. Azithromycin inhibits IL-1 secretion and non-canonical inflammasome activation. Sci Rep 2015; 5:12016. [PMID: 26152605 PMCID: PMC4495566 DOI: 10.1038/srep12016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/29/2015] [Indexed: 12/28/2022] Open
Abstract
Deregulation of inflammasome activation was recently identified to be involved in the pathogenesis of various inflammatory diseases. Although macrolide antibiotics display well described immunomodulatory properties, presumably involved in their clinical effects, their impact on inflammasome activation has not been investigated. We compared the influence of macrolides on cytokine induction in human monocytes. The role of intracellular azithromycin-accumulation was examined by interference with Ca++-dependent uptake. We have also analysed the signalling cascades involved in inflammasome activation, and substantiated the findings in a murine sepsis model. Azithromycin, but not clarithromycin or roxithromycin, specifically inhibited IL-1α and IL-1β secretion upon LPS stimulation. Interference with Ca++-dependent uptake abolished the cytokine-modulatory effect, suggesting a role of intracellular azithromycin accumulation in the modulatory role of this macrolide. Azithromycin’s inhibiting effects were observed upon LPS, but not upon flagellin, stimulation. Consistent with this observation, we found impaired induction of the LPS-sensing caspase-4 whereas NF-κB signalling was unaffected. Furthermore, azithromycin specifically affected IL-1β levels in a murine endotoxin sepsis model. We provide the first evidence of a differential impact of macrolides on the inflammasome/IL-1β axis, which may be of relevance in inflammasome-driven diseases such as chronic obstructive pulmonary disease or asthma.
Collapse
Affiliation(s)
- Guido A Gualdoni
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tilman Lingscheid
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Germany
| | - Klaus G Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Annika Hennig
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|