113 results on '"Andreas Wack"'
Search Results
2. Interplay between CXCR4 and CCR2 regulates bone marrow exit of dendritic cell progenitors
- Author
-
Mariana Pereira da Costa, Carlos M. Minutti, Cécile Piot, Evangelos Giampazolias, Ana Cardoso, Mar Cabeza-Cabrerizo, Neil C. Rogers, Marta Lebrusant-Fernandez, Chrysante S. Iliakis, Andreas Wack, and Caetano Reis e Sousa
- Subjects
CP: Developmental biology ,CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Conventional dendritic cells (cDCs) are found in most tissues and play a key role in initiation of immunity. cDCs require constant replenishment from progenitors called pre-cDCs that develop in the bone marrow (BM) and enter the blood circulation to seed all tissues. This process can be markedly accelerated in response to inflammation (emergency cDCpoiesis). Here, we identify two populations of BM pre-cDC marked by differential expression of CXCR4. We show that CXCR4lo cells constitute the migratory pool of BM pre-cDCs, which exits the BM and can be rapidly mobilized during challenge. We further show that exit of CXCR4lo pre-cDCs from BM at steady state is partially dependent on CCR2 and that CCR2 upregulation in response to type I IFN receptor signaling markedly increases efflux during infection with influenza A virus. Our results highlight a fine balance between retention and efflux chemokine cues that regulates steady-state and emergency cDCpoiesis.
- Published
- 2023
- Full Text
- View/download PDF
3. Hypoxia inducible factors regulate infectious SARS-CoV-2, epithelial damage and respiratory symptoms in a hamster COVID-19 model.
- Author
-
Peter A C Wing, Maria Prange-Barczynska, Amy Cross, Stefania Crotta, Claudia Orbegozo Rubio, Xiaotong Cheng, James M Harris, Xiaodong Zhuang, Rachel L Johnson, Kathryn A Ryan, Yper Hall, Miles W Carroll, Fadi Issa, Peter Balfe, Andreas Wack, Tammie Bishop, Francisco J Salguero, and Jane A McKeating
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Understanding the host pathways that define susceptibility to Severe-acute-respiratory-syndrome-coronavirus-2 (SARS-CoV-2) infection and disease are essential for the design of new therapies. Oxygen levels in the microenvironment define the transcriptional landscape, however the influence of hypoxia on virus replication and disease in animal models is not well understood. In this study, we identify a role for the hypoxic inducible factor (HIF) signalling axis to inhibit SARS-CoV-2 infection, epithelial damage and respiratory symptoms in the Syrian hamster model. Pharmacological activation of HIF with the prolyl-hydroxylase inhibitor FG-4592 significantly reduced infectious virus in the upper and lower respiratory tract. Nasal and lung epithelia showed a reduction in SARS-CoV-2 RNA and nucleocapsid expression in treated animals. Transcriptomic and pathological analysis showed reduced epithelial damage and increased expression of ciliated cells. Our study provides new insights on the intrinsic antiviral properties of the HIF signalling pathway in SARS-CoV-2 replication that may be applicable to other respiratory pathogens and identifies new therapeutic opportunities.
- Published
- 2022
- Full Text
- View/download PDF
4. Influenza A induces lactate formation to inhibit type I IFN in primary human airway epithelium
- Author
-
Jacob Thyrsted, Jacob Storgaard, Julia Blay-Cadanet, Alexander Heinz, Anne Laugaard Thielke, Stefania Crotta, Frank de Paoli, David Olagnier, Andreas Wack, Karsten Hiller, Anne Louise Hansen, and Christian Kanstrup Holm
- Subjects
Immune response ,Virology ,Metabolomics ,Science - Abstract
Summary: Pathogenic viruses induce metabolic changes in host cells to secure the availability of biomolecules and energy to propagate. Influenza A virus (IAV) and severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) both infect the human airway epithelium and are important human pathogens. The metabolic changes induced by these viruses in a physiologically relevant human model and how this affects innate immune responses to limit viral propagation are not well known. Using an ex vivo model of pseudostratified primary human airway epithelium, we here demonstrate that infection with both IAV and SARS-CoV-2 resulted in distinct metabolic changes including increases in lactate dehydrogenase A (LDHA) expression and LDHA-mediated lactate formation. Interestingly, LDHA regulated both basal and induced mitochondrial anti-viral signaling protein (MAVS)-dependent type I interferon (IFN) responses to promote IAV, but not SARS-CoV-2, replication. Our data demonstrate that LDHA and lactate promote IAV but not SARS-CoV-2 replication by inhibiting MAVS-dependent induction of type I IFN in primary human airway epithelium.
- Published
- 2021
- Full Text
- View/download PDF
5. Transcriptional profiling unveils type I and II interferon networks in blood and tissues across diseases
- Author
-
Akul Singhania, Christine M. Graham, Leona Gabryšová, Lúcia Moreira-Teixeira, Evangelos Stavropoulos, Jonathan M. Pitt, Probir Chakravarty, Annika Warnatsch, William J. Branchett, Laura Conejero, Jing-Wen Lin, Sophia Davidson, Mark S. Wilson, Gregory Bancroft, Jean Langhorne, Eva Frickel, Abdul K. Sesay, Simon L. Priestnall, Eleanor Herbert, Marianna Ioannou, Qian Wang, Ian R. Humphreys, Jonathan Dodd, Peter J. M. Openshaw, Katrin D. Mayer-Barber, Dragana Jankovic, Alan Sher, Clare M. Lloyd, Nicole Baldwin, Damien Chaussabel, Venizelos Papayannopoulos, Andreas Wack, Jacques F. Banchereau, Virginia M. Pascual, and Anne O’Garra
- Subjects
Science - Abstract
The authors present an extensive profile of host transcriptional respones to a diverse group of pathogens and allergens. In doing so, they identify TH1, type I IFN, TH17, and TH2 responses, that underlie each immune response in both the blood and lung, which represents a global profile of host-pathogen immune responses.
- Published
- 2019
- Full Text
- View/download PDF
6. Rotavirus susceptibility of antibiotic-treated mice ascribed to diminished expression of interleukin-22.
- Author
-
Daniel Schnepf, Pedro Hernandez, Tanel Mahlakõiv, Stefania Crotta, Meagan E Sullender, Stefan T Peterson, Annette Ohnemus, Camille Michiels, Ian Gentle, Laure Dumoutier, Celso A Reis, Andreas Diefenbach, Andreas Wack, Megan T Baldridge, and Peter Staeheli
- Subjects
Medicine ,Science - Abstract
The commensal microbiota regulates susceptibility to enteric pathogens by fine-tuning mucosal innate immune responses, but how susceptibility to enteric viruses is shaped by the microbiota remains incompletely understood. Past reports have indicated that commensal bacteria may either promote or repress rotavirus replication in the small intestine of mice. We now report that rotavirus replicated more efficiently in the intestines of germ-free and antibiotic-treated mice compared to animals with an unmodified microbiota. Antibiotic treatment also facilitated rotavirus replication in type I and type III interferon (IFN) receptor-deficient mice, revealing IFN-independent proviral effects. Expression of interleukin-22 (IL-22) was strongly diminished in the intestine of antibiotic-treated mice. Treatment with exogenous IL-22 blocked rotavirus replication in microbiota-depleted wild-type and Stat1-/- mice, demonstrating that the antiviral effect of IL-22 in animals with altered microbiome is not dependent on IFN signaling. In antibiotic-treated animals, IL-22-induced a specific set of genes including Fut2, encoding fucosyl-transferase 2 that participates in the biosynthesis of fucosylated glycans which can mediate rotavirus binding. Interestingly, IL-22 also blocked rotavirus replication in antibiotic-treated Fut2-/- mice. Furthermore, IL-22 inhibited rotavirus replication in antibiotic-treated mice lacking key molecules of the necroptosis or pyroptosis pathways of programmed cell death. Taken together, our results demonstrate that IL-22 determines rotavirus susceptibility of antibiotic-treated mice, yet the IL-22-induced effector molecules conferring rotavirus resistance remain elusive.
- Published
- 2021
- Full Text
- View/download PDF
7. Natural amines inhibit activation of human plasmacytoid dendritic cells through CXCR4 engagement
- Author
-
Nikaïa Smith, Nicolas Pietrancosta, Sophia Davidson, Jacques Dutrieux, Lise Chauveau, Pasquale Cutolo, Michel Dy, Daniel Scott-Algara, Bénédicte Manoury, Onofrio Zirafi, Isabelle McCort-Tranchepain, Thierry Durroux, Françoise Bachelerie, Olivier Schwartz, Jan Münch, Andreas Wack, Sébastien Nisole, and Jean-Philippe Herbeuval
- Subjects
Science - Abstract
Plasmacytoid dendritic cells produce type I interferons in response to viral sensing. Here the authors show that amines inhibit these plasmacytoid dendritic cell responses through CXCR4 engagement.
- Published
- 2017
- Full Text
- View/download PDF
8. Microbiota-Driven Tonic Interferon Signals in Lung Stromal Cells Protect from Influenza Virus Infection
- Author
-
Konrad C. Bradley, Katja Finsterbusch, Daniel Schnepf, Stefania Crotta, Miriam Llorian, Sophia Davidson, Serge Y. Fuchs, Peter Staeheli, and Andreas Wack
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Type I interferon (IFNα/β) pathways are fine-tuned to elicit antiviral protection while minimizing immunopathology; however, the initiating stimuli, target tissues, and underlying mechanisms are unclear. Using models of physiological and dysregulated IFNα/β receptor (IFNAR1) surface expression, we show here that IFNAR1-dependent signals set the steady-state IFN signature in both hematopoietic and stromal cells. Increased IFNAR1 levels promote a lung environment refractory to early influenza virus replication by elevating the baseline interferon signature. Commensal microbiota drive the IFN signature specifically in lung stroma, as shown by antibiotic treatment and fecal transplantation. Bone marrow chimera experiments identify lung stromal cells as crucially important for early antiviral immunity and stroma-immune cell interaction for late antiviral resistance. We propose that the microbiota-driven interferon signature in lung epithelia impedes early virus replication and that IFNAR1 surface levels fine-tune this signature. Our findings highlight the interplay between bacterial and viral exposure, with important implications for antibiotic use. : Bradley, Finsterbusch, et al. identify lung stroma as the target of microbiota-driven signals that set the interferon signature in these cells. Antibiotic treatment reduces gut microbiota and the lung stromal interferon signature and facilitates early influenza virus replication in lung epithelia, effects that can be reversed by fecal transplantation. Keywords: type I interferon (IFN), tonic IFN signaling, lung, microbiota, influenza, anti-viral protection, gut, antibiotics
- Published
- 2019
- Full Text
- View/download PDF
9. IFNλ is a potent anti‐influenza therapeutic without the inflammatory side effects of IFNα treatment
- Author
-
Sophia Davidson, Teresa M McCabe, Stefania Crotta, Hans Henrik Gad, Edith M Hessel, Soren Beinke, Rune Hartmann, and Andreas Wack
- Subjects
immunopathology ,infection ,influenza ,interferon alpha ,interferon lambda ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Influenza A virus (IAV)‐induced severe disease is characterized by infected lung epithelia, robust inflammatory responses and acute lung injury. Since type I interferon (IFNαβ) and type III interferon (IFNλ) are potent antiviral cytokines with immunomodulatory potential, we assessed their efficacy as IAV treatments. IFNλ treatment of IAV‐infected Mx1‐positive mice lowered viral load and protected from disease. IFNα treatment also restricted IAV replication but exacerbated disease. IFNα treatment increased pulmonary proinflammatory cytokine secretion, innate cell recruitment and epithelial cell death, unlike IFNλ‐treatment. IFNλ lacked the direct stimulatory activity of IFNα on immune cells. In epithelia, both IFNs induced antiviral genes but no inflammatory cytokines. Similarly, human airway epithelia responded to both IFNα and IFNλ by induction of antiviral genes but not of cytokines, while hPBMCs responded only to IFNα. The restriction of both IFNλ responsiveness and productive IAV replication to pulmonary epithelia allows IFNλ to limit IAV spread through antiviral gene induction in relevant cells without overstimulating the immune system and driving immunopathology. We propose IFNλ as a non‐inflammatory and hence superior treatment option for human IAV infection.
- Published
- 2016
- Full Text
- View/download PDF
10. The aryl hydrocarbon receptor controls cyclin O to promote epithelial multiciliogenesis
- Author
-
Matteo Villa, Stefania Crotta, Kevin S. Dingwell, Elizabeth M. A. Hirst, Manolis Gialitakis, Helena Ahlfors, James C. Smith, Brigitta Stockinger, and Andreas Wack
- Subjects
Science - Abstract
Epithelia are barriers against environmental insults and express the transcription factor aryl hydrocarbon receptor (AhR). Here the authors show that AhR regulates multiciliogenesis via cyclin O and Multicilin in a Notch-dependent manner and that this is blocked by toxic ligands.
- Published
- 2016
- Full Text
- View/download PDF
11. Interfering with transmission
- Author
-
Andreas Wack
- Subjects
Influenza virus ,interferons ,transmission ,respiratory tract ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The IFNλ family of interferons controls the spread of viruses in the upper respiratory tract and transmission between mice.
- Published
- 2018
- Full Text
- View/download PDF
12. Type I and type III interferons drive redundant amplification loops to induce a transcriptional signature in influenza-infected airway epithelia.
- Author
-
Stefania Crotta, Sophia Davidson, Tanel Mahlakoiv, Christophe J Desmet, Matthew R Buckwalter, Matthew L Albert, Peter Staeheli, and Andreas Wack
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.
- Published
- 2013
- Full Text
- View/download PDF
13. Contributors
- Author
-
Jamie Aranda, Goundappa K Balasubramani, Ritu Banerjee, Behin Barahimi, Laura A. Binari, David M. Brooks, Anna Burgner, Alexander De Castro-Abeger, Matthew Chinn, Amparo de la Peña, Ally Esch, Mark E. Garcia, Ben Geoffrey A.S., Pravin George, Karen E. Giles, Aarthi Goverdhan, Daniel Griffin, Sylvia Groth, Madihah Hepburn, Nancy Jacobson, Sarah Kim, Charles A. Knirsch, Srinivasan Krishnaswami, Sriram Krishnaswami, Pooja Lal, Susan M. Lopata, Subramani Mani, Natalie N. McCall, William M. McDonald, Sujatha S. Menon, Cullen P. Moran, Sivakumar Nagaraju, Christopher Newey, Mary Patricia Nowalk, Dhyanesh A. Patel, Amelie Pham, Krishna Ramakrishnamenon Prasad, Sathishkumar Ramalingam, John C. Ray, Mythily Srinivasan, Amit K. Srivastava, Jennifer L. Thompson, Thankam Thyvalikakath, Michael F. Vaezi, Andreas Wack, Robert S. Wallis, Charles B. Nemeroff, and Jörn-Hendrik Weitkamp
- Published
- 2024
14. Immunology of COVID-19
- Author
-
Robert Wallis, Amit K. Srivastava, Andreas Wack, and Charles A Knirsch
- Published
- 2024
15. Anti-type I interferon antibodies as a cause of severe COVID-19
- Author
-
ADRIAN HAYDAY, Gregory Towers, Ivan Zanoni, David Fajgenbaum, Angela Rogers, and Andreas Wack
- Subjects
Adult ,Aged, 80 and over ,Male ,SARS-CoV-2 ,Critical Illness ,Pneumonia, Viral ,COVID-19 ,Review Article ,Interferon alpha-2 ,Middle Aged ,Antibodies, Neutralizing ,Betacoronavirus ,Case-Control Studies ,Immunoglobulin G ,Interferon Type I ,Humans ,Female ,Coronavirus Infections ,Asymptomatic Infections ,Pandemics ,Aged ,Autoantibodies - Abstract
Interindividual clinical variability in the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is vast. We report that at least 101 of 987 patients with life-threatening coronavirus disease 2019 (COVID-19) pneumonia had neutralizing immunoglobulin G (IgG) autoantibodies (auto-Abs) against interferon-ω (IFN-ω) (13 patients), against the 13 types of IFN-α (36), or against both (52) at the onset of critical disease; a few also had auto-Abs against the other three type I IFNs. The auto-Abs neutralize the ability of the corresponding type I IFNs to block SARS-CoV-2 infection in vitro. These auto-Abs were not found in 663 individuals with asymptomatic or mild SARS-CoV-2 infection and were present in only 4 of 1227 healthy individuals. Patients with auto-Abs were aged 25 to 87 years and 95 of the 101 were men. A B cell autoimmune phenocopy of inborn errors of type I IFN immunity accounts for life-threatening COVID-19 pneumonia in at least 2.6% of women and 12.5% of men.
- Published
- 2022
16. Host-directed immunotherapy of viral and bacterial infections: past, present and future
- Author
-
Alan Sher, Andreas Wack, Anne O'Garra, and Robert S. Wallis
- Subjects
History ,Computer Science Applications ,Education - Abstract
The advent of COVID-19 and the persistent threat of infectious diseases such as tuberculosis, malaria, influenza and HIV/AIDS remind us of the marked impact that infections continue to have on public health. Some of the most effective protective measures are vaccines but these have been difficult to develop for some of these infectious diseases even after decades of research. The development of drugs and immunotherapies acting directly against the pathogen can be equally challenging, and such pathogen-directed therapeutics have the potential disadvantage of selecting for resistance. An alternative approach is provided by host-directed therapies, which interfere with host cellular processes required for pathogen survival or replication, or target the host immune response to infection (immunotherapies) to either augment immunity or ameliorate immunopathology. Here, we provide a historical perspective of host-directed immunotherapeutic interventions for viral and bacterial infections and then focus on SARS-CoV-2 and Mycobacterium tuberculosis, two major human pathogens of the current era, to indicate the key lessons learned and discuss candidate immunotherapeutic approaches, with a focus on drugs currently in clinical trials.
- Published
- 2022
17. Impaired immune response drives age-dependent severity of COVID-19
- Author
-
Julius Beer, Stefania Crotta, Angele Breithaupt, Annette Ohnemus, Jan Becker, Benedikt Sachs, Lisa Kern, Miriam Llorian, Nadine Ebert, Fabien Labroussaa, Tran Thi Nhu Thao, Bettina Salome Trueeb, Joerg Jores, Volker Thiel, Martin Beer, Jonas Fuchs, Georg Kochs, Andreas Wack, Martin Schwemmle, and Daniel Schnepf
- Subjects
Mice ,630 Agriculture ,SARS-CoV-2 ,Immunology ,Immunity ,Immunology and Allergy ,Animals ,COVID-19 ,630 Landwirtschaft ,Interferons ,Antiviral Agents - Abstract
SARS-CoV-2 is a highly contagious respiratory virus and the causative agent for COVID-19. The severity of disease varies from mildly symptomatic to lethal and shows an extraordinary correlation with increasing age, which represents the major risk factor for severe COVID-191. However, the precise pathomechanisms leading to aggravated disease in the elderly are currently unknown. Delayed and insufficient antiviral immune responses early after infection as well as dysregulated and overshooting immunopathological processes late during disease were suggested as possible mechanisms. Here we show that the age-dependent increase of COVID-19 severity is caused by the disruption of a timely and well-coordinated innate and adaptive immune response due to impaired interferon (IFN) responses. To overcome the limitations of mechanistic studies in humans, we generated a mouse model for severe COVID-19 and compared the kinetics of the immune responses in adult and aged mice at different time points after infection. Aggravated disease in aged mice was characterized by a diminished IFN-γ response and excessive virus replication. Accordingly, adult IFN-γ receptor-deficient mice phenocopied the age-related disease severity and supplementation of IFN-γ reversed the increased disease susceptibility of aged mice.Mimicking impaired type I IFN immunity in adult and aged mice, a second major risk factor for severe COVID-192–4, we found that therapeutic treatment with IFN-λ in adult and a combinatorial treatment with IFN-γ and IFN-λ in aged Ifnar1-/-mice was highly efficient in protecting against severe disease.Our findings provide an explanation for the age-dependent disease severity of COVID-19 and clarify the nonredundant antiviral functions of type I, II and III IFNs during SARS-CoV-2 infection in an age-dependent manner. Based on our data, we suggest that highly vulnerable individuals combining both risk factors, advanced age and an impaired type I IFN immunity, may greatly benefit from immunotherapy combining IFN-γ and IFN-λ.
- Published
- 2022
18. Tissue-specific and interferon-inducible expression of nonfunctional ACE2 through endogenous retroelement co-option
- Author
-
A.G. Wrobel, Andreas Wack, Jack Major, William Bolland, Jan Attig, Steve Gamblin, Kevin W. Ng, George R. Young, and George Kassiotis
- Subjects
Gene Expression Regulation, Viral ,Gene isoform ,Retroelements ,viruses ,Regulator ,Endogeny ,Stimulation ,Biology ,Gene Expression Regulation, Enzymologic ,Article ,Cell Line ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Interferon ,Chlorocebus aethiops ,Genetics ,medicine ,Animals ,Humans ,Tissue Distribution ,RNA-Seq ,Receptor ,Vero Cells ,030304 developmental biology ,0303 health sciences ,Protein Stability ,SARS-CoV-2 ,Gene Expression Profiling ,Translation (biology) ,Cell biology ,Isoenzymes ,HEK293 Cells ,Enzyme Induction ,Angiotensin-Converting Enzyme 2 ,Interferons ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,Receptors, Coronavirus ,medicine.drug - Abstract
Angiotensin-converting enzyme 2 (ACE2) is an entry receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and a regulator of several physiological processes. ACE2 has recently been proposed to be interferon-inducible, suggesting that SARS-CoV-2 may exploit this phenomenon to enhance viral spread and questioning the efficacy of interferon treatment in Coronavirus disease 2019 (COVID-19). Using a recent de novo transcript assembly that captured previously unannotated transcripts, we describe a novel isoform of ACE2, generated by co-option of intronic retroelements as promoter and alternative exon. The novel transcript, termed MIRb-ACE2, exhibits specific expression patterns across the aerodigestive and gastrointestinal tracts and is highly responsive to interferon stimulation. In stark contrast, canonical ACE2 expression is unresponsive to interferon stimulation. Moreover, the MIRb-ACE2 translation product is a truncated, unstable ACE2 form, lacking domains required for SARS-CoV-2 binding and is therefore unlikely to contribute to or enhance viral infection.
- Published
- 2020
19. Teaching Old Dogs New Tricks? The Plasticity of Lung Alveolar Macrophage Subsets
- Author
-
Justina Kulikauskaite and Andreas Wack
- Subjects
0301 basic medicine ,Opinion ,Cellular differentiation ,Cell Plasticity ,Immunology ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,Macrophages, Alveolar ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lung cancer ,Lung ,business.industry ,Monocyte ,respiratory system ,medicine.disease ,Embryonic stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Alveolar macrophage ,business ,Homeostasis ,030215 immunology - Abstract
Alveolar macrophages (AMs) are highly abundant lung cells with important roles in homeostasis and immunity. Their function influences the outcome of lung infections, lung cancer, and chronic inflammatory disease. Recent findings reveal functional heterogeneity of AMs. Following lung insult, resident AMs can either remain unchanged, acquire new functionality, or be replaced by monocyte-derived AMs. Evidence from mouse models correlates AM function with their embryonic or monocyte origin. We hypothesize that resident AMs are terminally differentiated cells with low responsiveness and limited plasticity, while recruited, monocyte-derived AMs are initially highly immunoreactive but more plastic, able to change their function in response to environmental cues. Understanding cell-intrinsic and -extrinsic mechanisms determining AM function may provide opportunities for intervention in lung disease., Highlights In mice at steady state, embryonically derived tissue-resident alveolar macrophages (AMs) self-sustain over a lifetime and fulfill their homeostatic function of surfactant removal. Following various lung insults, including influenza A virus infection and bleomycin-induced lung fibrosis, these cells show only minimal transcriptional and functional changes, suggesting that resident AMs are terminally differentiated, highly specialized, and not very plastic. In naïve mice, the contribution of monocyte-derived cells to AMs is negligible, but they become major components of the AM population post-lung insult. The functionality of recruited AMs can determine increased antibacterial protection, reduced asthma, and elevated lung fibrosis, depending on the specific initial insult studied. When the newly recruited population of monocyte-derived AMs persist in the mouse lung, they start to resemble tissue-resident AMs, suggesting that the lung environment sends cues to shape their relatively non-inflammatory, immunosedated phenotype. The high reactivity of recently recruited AMs might be best explained as a transcriptional and epigenetic legacy from their prior monocyte identity. Alternatively, training, acting through epigenetic and transcriptional reprogramming on hematopoietic stem cells or monocytes in the blood or in the lung, may imprint the high reactivity of recruited AMs.
- Published
- 2020
20. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection
- Author
-
Andreas Wack, Stefania Crotta, Rune Hartmann, Jack Major, Simon L. Priestnall, Miriam Llorian, Hans Henrik Gad, and Teresa M McCabe
- Subjects
Multidisciplinary ,Lung ,business.industry ,medicine.medical_treatment ,Cellular differentiation ,Endogeny ,medicine.disease_cause ,Proinflammatory cytokine ,Cytokine ,medicine.anatomical_structure ,Immunology ,medicine ,Influenza A virus ,Signal transduction ,business ,Interferon alfa ,medicine.drug - Abstract
Interferons interfere with lung repair Interferons (IFNs) are central to antiviral immunity. Viral recognition elicits IFN production, which in turn triggers the transcription of IFN-stimulated genes (ISGs), which engage in various antiviral functions. Type I IFNs (IFN-α and IFN-β) are widely expressed and can result in immunopathology during viral infections. By contrast, type III IFN (IFN-λ) responses are primarily restricted to mucosal surfaces and are thought to confer antiviral protection without driving damaging proinflammatory responses. Accordingly, IFN-λ has been proposed as a therapeutic in coronavirus disease 2019 (COVID-19) and other such viral respiratory diseases (see the Perspective by Grajales-Reyes and Colonna). Broggi et al. report that COVID-19 patient morbidity correlates with the high expression of type I and III IFNs in the lung. Furthermore, IFN-λ secreted by dendritic cells in the lungs of mice exposed to synthetic viral RNA causes damage to the lung epithelium, which increases susceptibility to lethal bacterial superinfections. Similarly, using a mouse model of influenza infection, Major et al. found that IFN signaling (especially IFN-λ) hampers lung repair by inducing p53 and inhibiting epithelial proliferation and differentiation. Complicating this picture, Hadjadj et al. observed that peripheral blood immune cells from severe and critical COVID-19 patients have diminished type I IFN and enhanced proinflammatory interleukin-6– and tumor necrosis factor-α–fueled responses. This suggests that in contrast to local production, systemic production of IFNs may be beneficial. The results of this trio of studies suggest that the location, timing, and duration of IFN exposure are critical parameters underlying the success or failure of therapeutics for viral respiratory infections. Science , this issue p. 706 , p. 712 , p. 718 ; see also p. 626
- Published
- 2020
21. A family of conserved bacterial virulence factors dampens interferon responses by blocking calcium signaling
- Author
-
Noémie Alphonse, Joseph J. Wanford, Andrew A. Voak, Jack Gay, Shayla Venkhaya, Owen Burroughs, Sanjana Mathew, Truelian Lee, Sasha L. Evans, Weiting Zhao, Kyle Frowde, Abrar Alrehaili, Ruth E. Dickenson, Mads Munk, Svetlana Panina, Ishraque F. Mahmood, Miriam Llorian, Megan L. Stanifer, Steeve Boulant, Martin W. Berchtold, Julien R.C. Bergeron, Andreas Wack, Cammie F. Lesser, and Charlotte Odendall
- Subjects
Model organisms ,Chemical Biology & High Throughput ,Human Biology & Physiology ,Virulence Factors ,FOS: Clinical medicine ,Genome Integrity & Repair ,Immunology ,Virulence Factors/metabolism ,Epithelial Cells ,Infectious Disease ,Tumour Biology ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Epithelial Cells/metabolism ,Mice ,Animals ,Interferons ,Calcium Signaling ,Genetics & Genomics ,Computational & Systems Biology ,Interferons/metabolism - Abstract
Interferons (IFNs) induce an antimicrobial state, protecting tissues from infection. Many viruses inhibit IFN signaling, but whether bacterial pathogens evade IFN responses remains unclear. Here, we demonstrate that the Shigella OspC family of type-III-secreted effectors blocks IFN signaling independently of its cell death inhibitory activity. Rather, IFN inhibition was mediated by the binding of OspC1 and OspC3 to the Ca2+ sensor calmodulin (CaM), blocking CaM kinase II and downstream JAK/STAT signaling. The growth of Shigella lacking OspC1 and OspC3 was attenuated in epithelial cells and in a murine model of infection. This phenotype was rescued in both models by the depletion of IFN receptors. OspC homologs conserved in additional pathogens not only bound CaM but also inhibited IFN, suggesting a widespread virulence strategy. These findings reveal a conserved but previously undescribed molecular mechanism of IFN inhibition and demonstrate the critical role of Ca2+ and IFN targeting in bacterial pathogenesis.
- Published
- 2022
22. SARS-CoV-2 S2-targeted vaccination elicits broadly neutralizing antibodies
- Author
-
Kevin W. Ng, Nikhil Faulkner, Katja Finsterbusch, Mary Wu, Ruth Harvey, Saira Hussain, Maria Greco, Yafei Liu, Svend Kjaer, Charles Swanton, Sonia Gandhi, Rupert Beale, Steve J. Gamblin, Peter Cherepanov, John McCauley, Rodney Daniels, Michael Howell, Hisashi Arase, Andreas Wack, David L.V. Bauer, and George Kassiotis
- Subjects
Model organisms ,COVID-19 Vaccines ,Immunology ,Infectious Disease ,Antibodies, Viral ,Biochemistry & Proteomics ,Coronavirus OC43, Human ,Mice ,Ecology,Evolution & Ethology ,Animals ,Humans ,Computational & Systems Biology ,Chemical Biology & High Throughput ,Human Biology & Physiology ,SARS-CoV-2 ,FOS: Clinical medicine ,Stem Cells ,Vaccination ,Genome Integrity & Repair ,Neurosciences ,COVID-19 ,General Medicine ,Cell Biology ,Tumour Biology ,Antibodies, Neutralizing ,Metabolism ,Spike Glycoprotein, Coronavirus ,Cell Cycle & Chromosomes ,Genetics & Genomics ,Broadly Neutralizing Antibodies ,Structural Biology & Biophysics - Abstract
Several variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged during the current coronavirus disease 2019 (COVID-19) pandemic. Although antibody cross-reactivity with the spike glycoproteins (S) of diverse coronaviruses, including endemic common cold coronaviruses (HCoVs), has been documented, it remains unclear whether such antibody responses, typically targeting the conserved S2 subunit, contribute to protection when induced by infection or through vaccination. Using a mouse model, we found that prior HCoV-OC43 S–targeted immunity primes neutralizing antibody responses to otherwise subimmunogenic SARS-CoV-2 S exposure and promotes S2-targeting antibody responses. Moreover, vaccination with SARS-CoV-2 S2 elicited antibodies in mice that neutralized diverse animal and human alphacoronaviruses and betacoronaviruses in vitro and provided a degree of protection against SARS-CoV-2 challenge in vivo. Last, in mice with a history of SARS-CoV-2 Wuhan–based S vaccination, further S2 vaccination induced broader neutralizing antibody response than booster Wuhan S vaccination, suggesting that it may prevent repertoire focusing caused by repeated homologous vaccination. These data establish the protective value of an S2-targeting vaccine and support the notion that S2 vaccination may better prepare the immune system to respond to the changing nature of the S1 subunit in SARS-CoV-2 variants of concern, as well as to future coronavirus zoonoses.
- Published
- 2022
- Full Text
- View/download PDF
23. Broad human and animal coronavirus neutralisation by SARS-CoV-2 S2-targeted vaccination
- Author
-
Peter Cherepanov, Yafei Liu, Hisashi Arase, Rodney S. Daniels, Kevin W. Ng, Katja Finsterbusch, Saira Hussain, Rupert Beale, Maria Greco, Svend Kjaer, David L.V. Bauer, John W. McCauley, Mary Wu, George Kassiotis, Michael Howell, Nikhil Faulkner, Andreas Wack, Ruth Harvey, Steve Gamblin, Charles Swanton, and Sonia Gandhi
- Subjects
chemistry.chemical_classification ,biology ,viruses ,virus diseases ,medicine.disease_cause ,Virology ,Neutralization ,Vaccination ,Immune system ,chemistry ,Immunity ,Pandemic ,medicine ,biology.protein ,Antibody ,Glycoprotein ,Coronavirus - Abstract
Several common-cold coronaviruses (HCoVs) are endemic in humans and several variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged during the current Coronavirus disease 2019 (COVID-19) pandemic. Whilst antibody cross-reactivity with the Spike glycoproteins (S) of diverse coronaviruses has been documented, it remains unclear whether such antibody responses, typically targeting the conserved S2 subunit, contribute to or mediate protection, when induced naturally or through vaccination. Using a mouse model, we show that prior HCoV-OC43 S immunity primes neutralising antibody responses to otherwise subimmunogenic SARS-CoV-2 S exposure and promotes S2-targeting antibody responses. Moreover, mouse vaccination with SARS-CoV-2 S2 elicits antibodies that neutralise diverse animal and human alphacoronaviruses and betacoronaviruses in vitro, and protects against SARS-CoV-2 challenge in vivo. Lastly, in mice with a history of SARS-CoV-2 Wuhan-based S vaccination, further S2 vaccination induces stronger and broader neutralising antibody response than booster Wuhan S vaccination, suggesting it may prevent repertoire focusing caused by repeated homologous vaccination. The data presented here establish the protective value of an S2-targeting vaccine and support the notion that S2 vaccination may better prepare the immune system to respond to the changing nature of the S1 subunit in SARS-CoV-2 variants of concern (VOCs), as well as to unpredictable, yet inevitable future coronavirus zoonoses.
- Published
- 2021
24. Influenza A induces lactate formation to inhibit type I IFN in primary human airway epithelium
- Author
-
Stefania Crotta, Christian K. Holm, Anne L. Thielke, Alexander Heinz, Julia Blay-Cadanet, Jacob Thyrsted, Karsten Hiller, Anne Louise Hansen, David Olagnier, Andreas Wack, Jacob Storgaard, and Frank Vincenzo de Paoli
- Subjects
Model organisms ,Science ,Lactate dehydrogenase A ,viruses ,Immunology ,Infectious Disease ,Biology ,medicine.disease_cause ,Virus ,Article ,Immune system ,Interferon ,Virology ,medicine ,Influenza A virus ,Metabolomics ,Respiratory system ,Immune response ,education ,skin and connective tissue diseases ,education.field_of_study ,Human Biology & Physiology ,Multidisciplinary ,Innate immune system ,FOS: Clinical medicine ,Epithelium ,Cell biology ,medicine.anatomical_structure ,medicine.drug - Abstract
Summary Pathogenic viruses induce metabolic changes in host cells to secure the availability of biomolecules and energy to propagate. Influenza A virus (IAV) and severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) both infect the human airway epithelium and are important human pathogens. The metabolic changes induced by these viruses in a physiologically relevant human model and how this affects innate immune responses to limit viral propagation are not well known. Using an ex vivo model of pseudostratified primary human airway epithelium, we here demonstrate that infection with both IAV and SARS-CoV-2 resulted in distinct metabolic changes including increases in lactate dehydrogenase A (LDHA) expression and LDHA-mediated lactate formation. Interestingly, LDHA regulated both basal and induced mitochondrial anti-viral signaling protein (MAVS)-dependent type I interferon (IFN) responses to promote IAV, but not SARS-CoV-2, replication. Our data demonstrate that LDHA and lactate promote IAV but not SARS-CoV-2 replication by inhibiting MAVS-dependent induction of type I IFN in primary human airway epithelium., Graphical abstract, Highlights • IAV and SARS-CoV-2 infections yield virus-specific changes in glucose metabolism • IAV and SARS-CoV-2 induce LDHA and lactate production in human airway epithelium • Lactate is highly pro-viral to IAV but not to SARS-CoV-2 • Lactate decreases IFN production and the following ISG response, Immune response; Virology; Metabolomics
- Published
- 2021
25. A TLR7 antagonist restricts interferon-dependent and -independent immunopathology in a mouse model of severe influenza
- Author
-
Matthias Mack, Stefania Crotta, Katja Finsterbusch, Andreas Wack, Julie C.F. Rappe, and Simon L. Priestnall
- Subjects
animal diseases ,medicine.medical_treatment ,Innate Immunity and Inflammation ,Immunology ,Inflammation ,Antiviral Agents ,Monocytes ,Infectious Disease and Host Defense ,Mice ,Immune system ,Orthomyxoviridae Infections ,Interferon ,Immunopathology ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lung ,Mice, Knockout ,Membrane Glycoproteins ,business.industry ,Macrophages ,Brief Definitive Report ,virus diseases ,Dendritic Cells ,Mucosal Immunology ,TLR7 ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Cytokine ,Toll-Like Receptor 7 ,Host-Pathogen Interactions ,Interferon Type I ,Cytokines ,medicine.symptom ,Cytokine storm ,business ,Interferon type I ,medicine.drug - Abstract
Distinct types of cytokine storms aggravate respiratory viral infection. This study shows that in severe influenza, TLR7 antagonist treatment ameliorates both type I interferon–driven and interferon-independent immunopathology by blocking TLR7-dependent cytokine production by immune cells, without affecting RIG-I–dependent responses in lung epithelia., Cytokine-mediated immune-cell recruitment and inflammation contribute to protection in respiratory virus infection. However, uncontrolled inflammation and the “cytokine storm” are hallmarks of immunopathology in severe infection. Cytokine storm is a broad term for a phenomenon with diverse characteristics and drivers, depending on host genetics, age, and other factors. Taking advantage of the differential use of virus-sensing systems by different cell types, we test the hypothesis that specifically blocking TLR7-dependent, immune cell–produced cytokines reduces influenza-related immunopathology. In a mouse model of severe influenza characterized by a type I interferon (IFN-I)–driven cytokine storm, TLR7 antagonist treatment leaves epithelial antiviral responses unaltered but acts through pDCs and monocytes to reduce IFN-I and other cytokines in the lung, thus ameliorating inflammation and severity. Moreover, even in the absence of IFN-I signaling, TLR7 antagonism reduces inflammation and mortality driven by monocyte-produced chemoattractants and neutrophil recruitment into the infected lung. Hence, TLR7 antagonism reduces diverse types of cytokine storm in severe influenza., Graphical Abstract
- Published
- 2021
26. Monocyte and dendritic cell defects in COVID-19
- Author
-
Andreas Wack
- Subjects
Coronavirus disease 2019 (COVID-19) ,Antigen presentation ,macromolecular substances ,Biology ,Antiviral Agents ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Interferon ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,SARS-CoV-2 ,Sequence Analysis, RNA ,Monocyte ,COVID-19 ,Cell Biology ,Dendritic cell ,Dendritic Cells ,Cell biology ,medicine.anatomical_structure ,Apoptosis ,030220 oncology & carcinogenesis ,Immunology ,medicine.drug - Abstract
Monocytes, plasmacytoid and conventional dendritic cells are crucial for antiviral immune responses. A new study now compares severe and moderate cases of COVID-19 and links defects in viral sensing, interferon and antigen presentation pathways, associated with upregulated apoptosis and inflammatory pathways, to high COVID-19 severity.
- Published
- 2021
27. Selective Janus kinase inhibition preserves interferon-λ-mediated antiviral responses
- Author
-
Celia Jakob, Juliane Vier, Laura Polcik, Stefania Crotta, Hans Henrik Gad, Miriam Llorian, Thiprampai Thamamongood, Rune Hartmann, Andreas Wack, Daniel Schnepf, Peter Staeheli, Birgit Strobl, Steeve Boulant, Annette Ohnemus, Martin Schwemmle, Susanne Kirschnek, and Megan L. Stanifer
- Subjects
Male ,0301 basic medicine ,Neutrophils ,medicine.medical_treatment ,Immunology ,Gene Expression ,Article ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Orthomyxoviridae Infections ,Heterocyclic Compounds ,Interferon ,medicine ,Animals ,Humans ,Janus Kinase Inhibitors ,Cells, Cultured ,Mice, Knockout ,TYK2 Kinase ,Sulfonamides ,TYK2 Deficiency ,Chemistry ,Epithelial Cells ,General Medicine ,Interferon λ ,030104 developmental biology ,Cytokine ,Influenza A virus ,Purines ,Tyrosine kinase 2 ,Cancer research ,Azetidines ,Pyrazoles ,Interferons ,Janus kinase ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Inflammatory diseases are frequently treated with Janus kinase (JAK) inhibitors to diminish cytokine signaling. These treatments can lead to inadvertent immune suppression and may increase the risk of viral infection. Tyrosine kinase 2 (TYK2) is a JAK family member required for efficient type I interferon (IFN-α/β) signaling. We report here that selective TYK2 inhibition preferentially blocked potentially detrimental type I IFN signaling, whereas IFN-λ-mediated responses were largely preserved. In contrast, the clinically used JAK1/2 inhibitor baricitinib was equally potent in blocking IFN-α/β- or IFN-λ-driven responses. Mechanistically, we showed that epithelial cells did not require TYK2 for IFN-λ-mediated signaling or antiviral protection. TYK2 deficiency diminished IFN-α- induced protection against lethal influenza virus infection in mice but did not impair IFN-λ-mediated antiviral protection. Our findings suggest that selective TYK2 inhibitors used in place of broadly acting JAK1/2 inhibitors may represent a superior treatment option for type I interferonopathies to counteract inflammatory responses while preserving antiviral protection mediated by IFN-λ.
- Published
- 2021
28. Estimating the effectiveness of routine asymptomatic PCR testing at different frequencies for the detection of SARS-CoV-2 infections
- Author
-
Dimitrios Evangelopoulos, Graeme Hewitt, Thomas Byrne, Kylie-ann Montgomery, Joshua Hope, Catherine Moore, Damien Tully, Jessica Diring, Pierre SANTUCCI, Paula Ordonez, Susan Michie, Christopher Jarvis, Carly Meyer, Abhinay Ramaprasad, Elise Crayton, Jernej Ule, Angela McBride, Margaux Silvestre, Joshua Wright, Steve Hindmarsh, Ed Manley, Johnathan Canton, Nina Vora, Efthymios Fidanis, Anabel Guedan, James MacRae, Gavin Kelly, Stefan Flasche, Mark Jit, Aaron Sait, Maryam Shahmanesh, Joel Hellewell, Andrew Riddell, Billy Quilty, Miriam Molina Arcas, Graham Medley, Claudio Bussi, Venizelos Papayannopoulos, Yang Liu, Matthew Quaife, Simon Procter, Hector Huerga Encabo, Rupert Beale, Christian Julian Villabona Arenas, Andreas Wack, Vascular Medicine, ACS - Pulmonary hypertension & thrombosis, and ACS - Atherosclerosis & ischemic syndromes
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Every other day ,Medical staff ,Care homes ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Presymptomatic infections ,Polymerase Chain Reaction ,Asymptomatic ,Bayes' theorem ,03 medical and health sciences ,0302 clinical medicine ,Pcr test ,Internal medicine ,medicine ,Humans ,Healthcare workers ,030212 general & internal medicine ,Symptom onset ,0303 health sciences ,SARS-CoV-2 ,030306 microbiology ,business.industry ,Test sensitivity ,COVID-19 ,Outbreak ,Bayes Theorem ,PCR testing ,General Medicine ,3. Good health ,030104 developmental biology ,COVID-19 Nucleic Acid Testing ,Medicine ,Female ,medicine.symptom ,business ,Research Article - Abstract
BackgroundRoutine asymptomatic testing using RT-PCR of people who interact with vulnerable populations, such as medical staff in hospitals or care workers in care homes, has been employed to help prevent outbreaks among vulnerable populations. Although the peak sensitivity of RT-PCR can be high, the probability of detecting an infection will vary throughout the course of an infection. The effectiveness of routine asymptomatic testing will therefore depend on testing frequency and how PCR detection varies over time.MethodsWe fitted a Bayesian statistical model to a dataset of twice weekly PCR tests of UK healthcare workers performed by self-administered nasopharyngeal swab, regardless of symptoms. We jointly estimated times of infection and the probability of a positive PCR test over time following infection, we then compared asymptomatic testing strategies by calculating the probability that a symptomatic infection is detected before symptom onset and the probability that an asymptomatic infection is detected within 7 days of infection.FindingsWe estimated that the probability that the PCR test detected infection peaked at 77% (54 - 88%) 4 days after infection, decreasing to 50% (38 - 65%) by 10 days after infection. Our results suggest a substantially higher probability of detecting infections 1–3 days after infection than previously published estimates. We estimated that testing every other day would detect 57% (33-76%) of symptomatic cases prior to onset and 94% (75-99%) of asymptomatic cases within 7 days if test results were returned within a day.InterpretationOur results suggest that routine asymptomatic testing can enable detection of a high proportion of infected individuals early in their infection, provided that the testing is frequent and the time from testing to notification of results is sufficiently fast.FundingWellcome Trust, National Institute for Health Research (NIHR) Health Protection Research Unit, Medical Research Council (UKRI)
- Published
- 2021
29. Severity of SARS-CoV-2 infection as a function of the interferon landscape across the respiratory tract of COVID-19 patients
- Author
-
Andrea Bottazzi, Alessandro Ambrosi, Roberto Ferrarese, Sofia Sisti, Antonio E. Pontiroli, Laura Saracino, Fabio A. Facchini, Benedetta Sposito, Laura Pandolfi, Andreas Wack, Laura Marongiu, Vanessa Frangipane, Massimo Clementi, Ivan Zanoni, Stefania Crotta, Enju Liu, Nicasio Mancini, Riccardo Colombo, Federica Meloni, Elena Tagliabue, Tommaso Fossali, Nicola Clementi, and Achille Broggi
- Subjects
Cell type ,medicine.anatomical_structure ,Coronavirus disease 2019 (COVID-19) ,Interferon ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,medicine ,Outbreak ,Biology ,Viral load ,Function (biology) ,Respiratory tract ,medicine.drug - Abstract
SummaryThe COVID-19 outbreak driven by SARS-CoV-2 has caused more than 2.5 million deaths globally, with the most severe cases characterized by over-exuberant production of immune-mediators, the nature of which is not fully understood. Interferons of the type I (IFN-I) or type III (IFN-III) families are potent antivirals, but their role in COVID-19 remains debated. Our analysis of gene and protein expression along the respiratory tract shows that IFNs, especially IFN-III, are over-represented in the lower airways of patients with severe COVID-19, while high levels of IFN-III, and to a lesser extent IFN-I, characterize the upper airways of patients with high viral burden but reduced disease risk or severity; also, IFN expression varies with abundance of the cell types that produce them. Our data point to a dynamic process of inter- and intra-family production of IFNs in COVID-19, and suggest that IFNs play opposing roles at distinct anatomical sites.
- Published
- 2021
30. Clinical outcomes of COVID-19 in long-term care facilities for people with epilepsy
- Author
-
Simona Balestrini, Matthias J. Koepp, Sonia Gandhi, Hannah M. Rickman, Gee Yen Shin, Catherine F. Houlihan, Jonny Anders-Cannon, Katri Silvennoinen, Fenglai Xiao, Sara Zagaglia, Kirsty Hudgell, Mariusz Ziomek, Paul Haimes, Adam Sampson, Annie Parker, J. Helen Cross, Rosemarie Pardington, Eleni Nastouli, Charles Swanton, Josemir W. Sander, Sanjay M. Sisodiya, Jim Aitken, Zoe Allen, Rachel Ambler, Karen Ambrose, Emma Ashton, Alida Avola, Samutheswari Balakrishnan, Caitlin Barns-Jenkins, Genevieve Barr, Sam Barrell, Souradeep Basu, Rupert Beale, Clare Beesley, Nisha Bhardwaj, Shahnaz Bibi, Ganka Bineva-Todd, Dhruva Biswas, Michael J. Blackman, Dominique Bonnet, Faye Bowker, Malgorzata Broncel, Claire Brooks, Michael D. Buck, Andrew Buckton, Timothy Budd, Alana Burrell, Louise Busby, Claudio Bussi, Simon Butterworth, Matthew Byott, Fiona Byrne, Richard Byrne, Simon Caidan, Joanna Campbell, Johnathan Canton, Ana Cardoso, Nick Carter, Luiz Carvalho, Raffaella Carzaniga, Natalie Chandler, Qu Chen, Peter Cherepanov, Laura Churchward, Graham Clark, Bobbi Clayton, Clementina Cobolli Gigli, Zena Collins, Sally Cottrell, Margaret Crawford, Laura Cubitt, Tom Cullup, Heledd Davies, Patrick Davis, Dara Davison, Vicky Dearing, Solene Debaisieux, Monica Diaz-Romero, Alison Dibbs, Jessica Diring, Paul C. Driscoll, Annalisa D'Avola, Christopher Earl, Amelia Edwards, Chris Ekin, Dimitrios Evangelopoulos, Rupert Faraway, Antony Fearns, Aaron Ferron, Efthymios Fidanis, Dan Fitz, James Fleming, Daniel Frampton, Bruno Frederico, Alessandra Gaiba, Anthony Gait, Steve Gamblin, Kathleen Gärtner, Liam Gaul, Helen M. Golding, Jacki Goldman, Robert Goldstone, Belen Gomez Dominguez, Hui Gong, Paul R. Grant, Maria Greco, Mariana Grobler, Anabel Guedan, Maximiliano G. Gutierrez, Fiona Hackett, Ross Hall, Steinar Halldorsson, Suzanne Harris, Sugera Hashim, Emine Hatipoglu, Lyn Healy, Judith Heaney, Susanne Herbst, Graeme Hewitt, Theresa Higgins, Steve Hindmarsh, Rajnika Hirani, Joshua Hope, Elizabeth Horton, Beth Hoskins, Michael Howell, Louise Howitt, Jacqueline Hoyle, Mint R. Htun, Michael Hubank, Hector Huerga Encabo, Deborah Hughes, Jane Hughes, Almaz Huseynova, Ming-Shih Hwang, Rachael Instrell, Deborah Jackson, Mariam Jamal-Hanjani, Lucy Jenkins, Ming Jiang, Mark Johnson, Leigh Jones, Nnennaya Kanu, George Kassiotis, Gavin Kelly, Louise Kiely, Anastacio King Spert Teixeira, Stuart Kirk, Svend Kjaer, Ellen Knuepfer, Nikita Komarov, Paul Kotzampaltiris, Konstantinos Kousis, Tammy Krylova, Ania Kucharska, Robyn Labrum, Catherine Lambe, Michelle Lappin, Stacey-Ann Lee, Andrew Levett, Lisa Levett, Marcel Levi, Hon Wing Liu, Sam Loughlin, Wei-Ting Lu, James I. MacRae, Akshay Madoo, Julie A. Marczak, Mimmi Martensson, Thomas Martinez, Bishara Marzook, John Matthews, Joachim M. Matz, Samuel McCall, Laura E. McCoy, Fiona McKay, Edel C. McNamara, Carlos M. Minutti, Gita Mistry, Miriam Molina-Arcas, Beatriz Montaner, Kylie Montgomery, Catherine Moore, David Moore, Anastasia Moraiti, Lucia Moreira-Teixeira, Joyita Mukherjee, Cristina Naceur-Lombardelli, Aileen Nelson, Jerome Nicod, Luke Nightingale, Stephanie Nofal, Paul Nurse, Savita Nutan, Caroline Oedekoven, Anne O'Garra, Jean D. O'Leary, Jessica Olsen, Olga O'Neill, Nicola O'Reilly, Paula Ordonez Suarez, Neil Osborne, Amar Pabari, Aleksandra Pajak, Venizelos Papayannopoulos, Stavroula M Paraskevopoulou, Namita Patel, Yogen Patel, Oana Paun, Nigel Peat, Laura Peces-Barba Castano, Ana Perez Caballero, Jimena Perez-Lloret, Magali S. Perrault, Abigail Perrin, Roy Poh, Enzo Z. Poirier, James M. Polke, Marc Pollitt, Lucia Prieto-Godino, Alize Proust, Clinda Puvirajasinghe, Christophe Queval, Vijaya Ramachandran, Abhinay Ramaprasad, Peter Ratcliffe, Laura Reed, Caetano Reis e Sousa, Kayleigh Richardson, Sophie Ridewood, Fiona Roberts, Rowenna Roberts, Angela Rodgers, Pablo Romero Clavijo, Annachiara Rosa, Alice Rossi, Chloe Roustan, Andrew Rowan, Erik Sahai, Aaron Sait, Katarzyna Sala, Emilie Sanchez, Theo Sanderson, Pierre Santucci, Fatima Sardar, Adam Sateriale, Jill A. Saunders, Chelsea Sawyer, Anja Schlott, Edina Schweighoffer, Sandra Segura-Bayona, Rajvee Shah Punatar, Maryam Shahmanesh, Joe Shaw, Mariana Silva Dos Santos, Margaux Silvestre, Matthew Singer, Daniel M. Snell, Ok-Ryul Song, Moira J. Spyer, Louisa Steel, Amy Strange, Adrienne E. Sullivan, Michele S.Y. Tan, Zoe H. Tautz-Davis, Effie Taylor, Gunes Taylor, Harriet B. Taylor, Alison Taylor-Beadling, Fernanda Teixeira Subtil, Berta Terré Torras, Patrick Toolan-Kerr, Francesca Torelli, Tea Toteva, Moritz Treeck, Hadija Trojer, Ming-Han C. Tsai, James M.A. Turner, Melanie Turner, Jernej Ule, Rachel Ulferts, Sharon P. Vanloo, Selvaraju Veeriah, Subramanian Venkatesan, Karen Vousden, Andreas Wack, Claire Walder, Philip A. Walker, Yiran Wang, Sophia Ward, Catharina Wenman, Luke Williams, Matthew J. Williams, Wai Keong Wong, Joshua Wright, Mary Wu, Lauren Wynne, Zheng Xiang, Melvyn Yap, Julian A. Zagalak, Davide Zecchin, Rachel Zillwood, Santhakumari Carthiyaniamma, Jane DeTisi, Julie Dick, Andrea Hill, Karin Kipper, Birinder Kullar, Sarah Norris, Fergus Rugg-Gunn, Rebecca Salvatierra, Gabriel Shaya, Astrid Sloan, Priyanka Singh, James Varley, Ben Whatley, and Academic Medical Center
- Subjects
Male ,Pediatrics ,CCC, Crick COVID Consortium ,SWGC, Sir William Gowers Centre ,Comorbidity ,Residential Facilities ,Cohort Studies ,Behavioral Neuroscience ,Epilepsy ,0302 clinical medicine ,Case fatality rate ,Infection control ,030212 general & internal medicine ,Care Models ,COVID-19, coronavirus disease 2019 ,Aged, 80 and over ,Surveillance ,Middle Aged ,3. Good health ,Treatment Outcome ,Neurology ,UCLH, University College London Hospitals NHS Foundation Trust ,PEG, percutaneous endoscopic gastrostomy ,STE, St Elizabeth’s Centre ,Female ,medicine.symptom ,PPE, personal protective equipment ,Cohort study ,Adult ,medicine.medical_specialty ,Isolation (health care) ,Clinical Neurology ,Asymptomatic ,Article ,Young Adult ,03 medical and health sciences ,medicine ,Humans ,YE, Young Epilepsy ,Aged ,SARS-CoV-2, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Infection Control ,business.industry ,SARS-CoV-2 ,Prevention ,Vulnerable people ,COVID-19 ,TM, The Meath ,medicine.disease ,Long-Term Care ,CCE, Chalfont Centre for Epilepsy ,United Kingdom ,Long-term care ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Contact tracing - Abstract
Highlights • We found a high asymptomatic rate in vulnerable people with epilepsy. • Enhanced surveillance allows to quickly contain outbreaks. • We report a low rate of COVID-19 morbidity and mortality in a long-term care facility. • Preventative measures allow reducing resident-to-resident and -to-caregiver transmission. • Children and young adults appear to have lower infection rates., In this cohort study, we aim to compare outcomes from coronavirus disease 2019 (COVID-19) in people with severe epilepsy and other co-morbidities living in long-term care facilities which all implemented early preventative measures, but different levels of surveillance. During 25-week observation period (16 March–6 September 2020), we included 404 residents (118 children), and 1643 caregivers. We compare strategies for infection prevention, control, and containment, and related outcomes, across four UK long-term care facilities. Strategies included early on-site enhancement of preventative and infection control measures, early identification and isolation of symptomatic cases, contact tracing, mass surveillance of asymptomatic cases and contacts. We measured infection rate among vulnerable people living in the facilities and their caregivers, with asymptomatic and symptomatic cases, including fatality rate. We report 38 individuals (17 residents) who tested severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-positive, with outbreaks amongst residents in two facilities. At Chalfont Centre for Epilepsy (CCE), 10/98 residents tested positive: two symptomatic (one died), eight asymptomatic on weekly enhanced surveillance; 2/275 caregivers tested positive: one symptomatic, one asymptomatic. At St Elizabeth’s (STE), 7/146 residents tested positive: four symptomatic (one died), one positive during hospital admission for symptoms unrelated to COVID-19, two asymptomatic on one-off testing of all 146 residents; 106/601 symptomatic caregivers were tested, 13 positive. In addition, during two cycles of systematically testing all asymptomatic carers, four tested positive. At The Meath (TM), 8/80 residents were symptomatic but none tested; 26/250 caregivers were tested, two positive. At Young Epilepsy (YE), 8/80 children were tested, all negative; 22/517 caregivers were tested, one positive. Infection outbreaks in long-term care facilities for vulnerable people with epilepsy can be quickly contained, but only if asymptomatic individuals are identified through enhanced surveillance at resident and caregiver level. We observed a low rate of morbidity and mortality, which confirmed that preventative measures with isolation of suspected and confirmed COVID-19 residents can reduce resident-to-resident and resident-to-caregiver transmission. Children and young adults appear to have lower infection rates. Even in people with epilepsy and multiple co-morbidities, we observed a high percentage of asymptomatic people suggesting that epilepsy-related factors (anti-seizure medications and seizures) do not necessarily lead to poor outcomes.
- Published
- 2021
31. Influenza A Virus Induces LDHA Expression and Lactate Formation to Inhibit Type I IFN and Promote Replication in Primary Human Airway Epithelium
- Author
-
Andreas Wack, Christian K. Holm, Julia Blay-Cadanet, Stefania Crotta, Jacob Thyrsted, David Olagnier, Karsten Hiller, Jacob Storgaard, Anne Louise Hansen, Anne L. Thielke, Frank Vincenzo de Paoli, and Alexander Heinz
- Subjects
education.field_of_study ,Innate immune system ,Lactate dehydrogenase A ,Biology ,medicine.disease_cause ,Virology ,Epithelium ,medicine.anatomical_structure ,Interferon ,medicine ,Influenza A virus ,Respiratory epithelium ,Glycolysis ,education ,Ex vivo ,medicine.drug - Abstract
Pathogenic viruses induce metabolic changes in host cells to secure availability of biomolecules and energy to propagate. Influenza A virus and SARS-CoV-2 both infect the human airway epithelium and are important human pathogens. The metabolic changes induced by these viruses in a physiologically relevant human model, and how this affects innate immune responses to limit viral propagation is not well known. Using an ex vivo model of pseudostratified primary human airway epithelium, we here demonstrate that infection with both IAV and SARS-CoV-2 resulted in distinct metabolic changes including increases in lactate Dehydrogenase A (LDHA) expression and LDHA-mediated lactate formation. Interestingly, LDHA regulated both basal and induced MAVS-dependent type I interferon (IFN) responses to promote IAV, but not SARS-CoV-2, replication. Our data demonstrate that LDHA and Lactate promote IAV but not SARS-CoV-2 replication by inhibiting MAVS-dependent induction of type I IFN in primary human airway epithelium. Funding Statement: This research work was supported by the Graduate School of Health Aarhus University, Fhv. Dir. Leo Nielsen & Hustru Karen Margrethe Nielsens Legat for Laegevidenskabelig Grundforskning, Ester M. & Konrad Kristian Sigurdssons Dyrevaernsfond, Beckett-fonden, Kong Christian IX & Dronning Louises Jubilaeumslegat, Christian Larsen & Dommer Ellen Larsens Legat, Direktor Emil C. Hertz & hustru Inger Hertz’ fond og A.P. Moller Fonden. SC and AW were supported by the Francis Crick Institute which receives its core funding from Cancer Research UK (FC001206), the UK Medical Council (FC001206), and the Wellcome Trust (FC001206). Declaration of Interests: The authors declare no competing interests.
- Published
- 2021
32. The interferon landscape along the respiratory tract impacts the severity of COVID-19
- Author
-
Riccardo Colombo, Ivan Zanoni, Andreas Wack, Achille Broggi, Nicola Clementi, Tommaso Fossali, Laura Marongiu, Elena Tagliabue, Andrea Bottazzi, Fabio A. Facchini, Sofia Sisti, Janet Chou, Roberto Spreafico, Roberto Ferrarese, Elena Criscuolo, Vanessa Frangipane, Jaclyn M. Long, Federica Meloni, Nicasio Mancini, Sara Bozzini, Stefania Crotta, Benedetta Sposito, Massimo Clementi, Enju Liu, Antonio E. Pontiroli, Laura Pandolfi, Alessandro Ambrosi, Laura Saracino, Harvard Medical School [Boston] (HMS), Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Pavia = University of Pavia (UNIPV), The Francis Crick Institute [London], Universita Vita Salute San Raffaele = Vita-Salute San Raffaele University [Milan, Italie] (UniSR), Institute for Quantitative and Computational Biosciences - University of california LA, Division of Gastroenterology [Boston, MA, USA], Harvard University-Harvard Medical School [Boston] (HMS)-Beth Israel Deaconess Medical Center [Boston] (BIDMC), Dipartimento di Informatica, Matematica, Elettronica e Trasporti [Reggio Calabria] (DIMET), Universita Mediterranea of Reggio Calabria [Reggio Calabria], Luigi sacco hospital - Division of Anesthesiology and intensive Care, IRCCS Multimedica, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, Università degli Studi di Milano = University of Milan (UNIMI), Division of Respiratory Diseases (IRCCS), Università degli Studi di Pavia = University of Pavia (UNIPV)-San Matteo' Hospital, IRCCS laboratory of medical microbiology and virology Milan, Harvard University [Cambridge]-Harvard Medical School [Boston] (HMS)-Beth Israel Deaconess Medical Center [Boston] (BIDMC), DUMENIL, Anita, Sposito, B, Broggi, A, Pandolfi, L, Crotta, S, Clementi, N, Ferrarese, R, Sisti, S, Criscuolo, E, Spreafico, R, Long, J, Ambrosi, A, Liu, E, Frangipane, V, Saracino, L, Bozzini, S, Marongiu, L, Facchini, F, Bottazzi, A, Fossali, T, Colombo, R, Clementi, M, Tagliabue, E, Chou, J, Pontiroli, A, Meloni, F, Wack, A, Mancini, N, Zanoni, I, Sposito, Benedetta, Broggi, Achille, Pandolfi, Laura, Crotta, Stefania, Clementi, Nicola, Ferrarese, Roberto, Sisti, Sofia, Criscuolo, Elena, Spreafico, Roberto, Long, Jaclyn M., Ambrosi, Alessandro, Liu, Enju, Frangipane, Vanessa, Saracino, Laura, Bozzini, Sara, Marongiu, Laura, Facchini, Fabio A., Bottazzi, Andrea, Fossali, Tommaso, Colombo, Riccardo, Clementi, Massimo, Tagliabue, Elena, Chou, Janet, Pontiroli, Antonio E., Meloni, Federica, Wack, Andrea, Mancini, Nicasio, and Zanoni, Ivan
- Subjects
Model organisms ,Aging ,dendritic cell ,pattern recognition receptor ,[SDV]Life Sciences [q-bio] ,Respiratory System ,Immunology ,Type I IFN ,Infectious Disease ,Biology ,Severity of Illness Index ,Article ,General Biochemistry, Genetics and Molecular Biology ,lung ,Interferon ,Type III IFN ,Severity of illness ,Leukocytes ,medicine ,Humans ,Respiratory system ,ComputingMilieux_MISCELLANEOUS ,Regulation of gene expression ,Respiratory Distress Syndrome ,Human Biology & Physiology ,Lung ,SARS-CoV-2 ,epithelial cell ,FOS: Clinical medicine ,Age Factors ,Pattern recognition receptor ,COVID-19 ,Epithelial Cells ,interferon ,Viral Load ,COVID-19, SARS-CoV-2, Type I IFN, Type III IFN, airways, dendritic cell, epithelial cell, interferon, lung ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Gene Expression Regulation ,airways ,airway ,Interferons ,Viral load ,Respiratory tract ,medicine.drug - Abstract
Severe COVID-19 is characterized by overproduction of immune mediators, but the role of interferons (IFNs) of the type I (IFN-I) or type III (IFN-III) families remains debated. We scrutinized the production of IFNs along the respiratory tract of COVID-19 patients and found that high levels of IFN-III, and to a lesser extent IFN-I, characterize the upper airways of patients with high viral burden but reduced disease risk or severity. Production of specific IFN-III, but not IFN-I, members, denotes patients with a mild pathology and efficiently drives the transcription of genes that protect against SARS-CoV-2. In contrast, compared to subjects with other infectious or non-infectious lung pathologies, IFNs are over-represented in the lower airways of patients with severe COVID-19 that exhibit gene pathways associated with increased apoptosis and decreased proliferation. Our data demonstrate a dynamic production of IFNs in SARS-CoV-2-infected patients and show IFNs play opposing roles at distinct anatomical sites., An in-depth analysis of interferons in COVID-19 reveals differences in their roles based on anatomical location, viral load, age and disease severity. In the upper respiratory tract, high levels of IFN-III are protective and result in mild disease in spite of higher SARS-CoV-2 viral burden while the lower airways of patients with severe COVID-19 demonstrate elevated IFN-I and III, cell death and a reduction in interferon stimulated genes.
- Published
- 2021
- Full Text
- View/download PDF
33. Tissue-specific and interferon-inducible expression of non-functional ACE2 through endogenous retrovirus co-option
- Author
-
Jack Major, William Bolland, Andreas Wack, Kevin W. Ng, Jan Attig, George R. Young, and George Kassiotis
- Subjects
Gene isoform ,Interferon ,Regulator ,medicine ,Endogenous retrovirus ,Translation (biology) ,Stimulation ,Biology ,Receptor ,hormones, hormone substitutes, and hormone antagonists ,Long terminal repeat ,medicine.drug ,Cell biology - Abstract
SummaryAngiotensin-converting enzyme 2 (ACE2) is an entry receptor for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), as well as a regulator of several physiological processes. ACE2 has recently been proposed to be interferon-inducible, suggesting that SARS-CoV-2 may exploit this phenomenon to enhance viral spread and questioning the efficacy of interferon treatment in Coronavirus disease 2019 (COVID-19). Using a recent de novo transcript assembly that captured previously unannotated transcripts, we describe a novel isoform of ACE2, generated by co-option of an intronic long terminal repeat (LTR) retroelement promoter. The novel transcript, termed LTR16A1-ACE2, exhibits specific expression patterns across the aerodigestive and gastrointestinal tracts and, importantly, is highly responsive to interferon stimulation. In stark contrast, expression of canonical ACE2 is completely unresponsive to interferon stimulation. Moreover, the LTR16A1-ACE2 translation product is a truncated, unstable ACE2 form, lacking domains required for SARS-CoV-2 binding and therefore unlikely to contribute to or enhance viral infection.
- Published
- 2020
34. Pandemic peak SARS-CoV-2 infection and seroconversion rates in London frontline health-care workers
- Author
-
Catherine F Houlihan, Nina Vora, Thomas Byrne, Dan Lewer, Gavin Kelly, Judith Heaney, Sonia Gandhi, Moira J Spyer, Rupert Beale, Peter Cherepanov, David Moore, Richard Gilson, Steve Gamblin, George Kassiotis, Laura E McCoy, Charles Swanton, Andrew Hayward, Eleni Nastouli, Jim Aitken, Zoe Allen, Rachel Ambler, Karen Ambrose, Emma Ashton, Alida Avola, Samutheswari Balakrishnan, Caitlin Barns-Jenkins, Genevieve Barr, Sam Barrell, Souradeep Basu, Clare Beesley, Nisha Bhardwaj, Shahnaz Bibi, Ganka Bineva-Todd, Dhruva Biswas, Michael J Blackman, Dominique Bonnet, Faye Bowker, Malgorzata Broncel, Claire Brooks, Michael D Buck, Andrew Buckton, Timothy Budd, Alana Burrell, Louise Busby, Claudio Bussi, Simon Butterworth, Fiona Byrne, Richard Byrne, Simon Caidan, Joanna Campbell, Johnathan Canton, Ana Cardoso, Nick Carter, Luiz Carvalho, Raffaella Carzaniga, Natalie Chandler, Qu Chen, Laura Churchward, Graham Clark, Bobbi Clayton, Clementina Cobolli Gigli, Zena Collins, Sally Cottrell, Margaret Crawford, Laura Cubitt, Tom Cullup, Heledd Davies, Patrick Davis, Dara Davison, Annalisa D'Avola, Vicky Dearing, Solene Debaisieux, Monica Diaz-Romero, Alison Dibbs, Jessica Diring, Paul C Driscoll, Christopher Earl, Amelia Edwards, Chris Ekin, Dimitrios Evangelopoulos, Rupert Faraway, Antony Fearns, Aaron Ferron, Efthymios Fidanis, Dan Fitz, James Fleming, Bruno Frederico, Alessandra Gaiba, Anthony Gait, Liam Gaul, Helen M Golding, Jacki Goldman, Robert Goldstone, Belen Gomez Dominguez, Hui Gong, Paul R Grant, Maria Greco, Mariana Grobler, Anabel Guedan, Maximiliano G Gutierrez, Fiona Hackett, Ross Hall, Steinar Halldorsson, Suzanne Harris, Sugera Hashim, Lyn Healy, Susanne Herbst, Graeme Hewitt, Theresa Higgins, Steve Hindmarsh, Rajnika Hirani, Joshua Hope, Elizabeth Horton, Beth Hoskins, Michael Howell, Louise Howitt, Jacqueline Hoyle, Mint R Htun, Michael Hubank, Hector Huerga Encabo, Deborah Hughes, Jane Hughes, Almaz Huseynova, Ming-Shih Hwang, Rachael Instrell, Deborah Jackson, Mariam Jamal-Hanjani, Lucy Jenkins, Ming Jiang, Mark Johnson, Leigh Jones, Nnennaya Kanu, Louise Kiely, Anastacio King Spert Teixeira, Stuart Kirk, Svend Kjaer, Ellen Knuepfer, Nikita Komarov, Paul Kotzampaltiris, Konstantinos Kousis, Tammy Krylova, Ania Kucharska, Robyn Labrum, Catherine Lambe, Michelle Lappin, Stacey-Ann Lee, Andrew Levett, Lisa Levett, Marcel Levi, Hon-Wing Liu, Sam Loughlin, Wei-Ting Lu, James I MacRae, Akshay Madoo, Julie A Marczak, Mimmi Martensson, Thomas Martinez, Bishara Marzook, John Matthews, Joachim M Matz, Samuel McCall, Fiona McKay, Edel C McNamara, Carlos M Minutti, Gita Mistry, Miriam Molina-Arcas, Beatriz Montaner, Kylie Montgomery, Catherine Moore, Anastasia Moraiti, Lucia Moreira-Teixeira, Joyita Mukherjee, Cristina Naceur-Lombardelli, Aileen Nelson, Jerome Nicod, Luke Nightingale, Stephanie Nofal, Paul Nurse, Savita Nutan, Caroline Oedekoven, Anne O'Garra, Jean D O'Leary, Jessica Olsen, Olga O'Neill, Paula Ordonez Suarez, Nicola O'Reilly, Neil Osborne, Amar Pabari, Aleksandra Pajak, Venizelos Papayannopoulos, Namita Patel, Yogen Patel, Oana Paun, Nigel Peat, Laura Peces-Barba Castano, Ana Perez Caballero, Jimena Perez-Lloret, Magali S Perrault, Abigail Perrin, Roy Poh, Enzo Z Poirier, James M Polke, Marc Pollitt, Lucia Prieto-Godino, Alize Proust, Rajvee Shah Punatar, Clinda Puvirajasinghe, Christophe Queval, Vijaya Ramachandran, Abhinay Ramaprasad, Peter Ratcliffe, Laura Reed, Caetano Reis e Sousa, Kayleigh Richardson, Sophie Ridewood, Rowenna Roberts, Angela Rodgers, Pablo Romero Clavijo, Annachiara Rosa, Alice Rossi, Chloe Roustan, Andrew Rowan, Erik Sahai, Aaron Sait, Katarzyna Sala, Theo Sanderson, Pierre Santucci, Fatima Sardar, Adam Sateriale, Jill A Saunders, Chelsea Sawyer, Anja Schlott, Edina Schweighoffer, Sandra Segura-Bayona, Joe Shaw, Gee Yen Shin, Mariana Silva Dos Santos, Margaux Silvestre, Matthew Singer, Daniel M Snell, Ok-Ryul Song, Louisa Steel, Amy Strange, Adrienne E Sullivan, Michele SY Tan, Zoe H Tautz-Davis, Effie Taylor, Gunes Taylor, Harriet B Taylor, Alison Taylor-Beadling, Fernanda Teixeira Subtil, Berta Terré Torras, Patrick Toolan-Kerr, Francesca Torelli, Tea Toteva, Moritz Treeck, Hadija Trojer, Ming-Han C Tsai, James MA Turner, Melanie Turner, Jernej Ule, Rachel Ulferts, Sharon P Vanloo, Selvaraju Veeriah, Subramanian Venkatesan, Karen Vousden, Andreas Wack, Claire Walder, Philip A Walker, Yiran Wang, Sophia Ward, Catharina Wenman, Luke Wiliams, Matthew J Williams, Wai Keong Wong, Joshua Wright, Mary Wu, Lauren Wynne, Zheng Xiang, Melvyn Yap, Julian A Zagalak, Davide Zecchin, Rachel Zillwood, Rebecca Matthews, Abigail Severn, Sajida Adam, Louise Enfield, Angela McBride, Kathleen Gärtner, Sarah Edwards, Fabiana Lorencatto, Susan Michie, Ed Manley, Maryam Shahmanesh, Hinal Lukha, Paulina Prymas, Hazel McBain, Robert Shortman, Leigh Wood, Claudia Davies, Bethany Williams, Kevin W Ng, Georgina H Cornish, Nikhil Faulkner, Andrew Riddell, Philip Hobson, Ana Agua-Doce, Kerol Bartolovic, Emma Russell, Lotte Carr, Emilie Sanchez, Daniel Frampton, Matthew Byott, Stavroula M Paraskevopoulou, Elise Crayton, Carly Meyer, Triantafylia Gkouleli, Andrea Stoltenberg, Veronica Ranieri, Tom Byrne, Fiona Roberts, and Emine Hatipoglu
- Subjects
Adult ,2019-20 coronavirus outbreak ,Infectious Disease Transmission, Patient-to-Professional ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Health Personnel ,Pneumonia, Viral ,Viral transmission ,Antibodies, Viral ,Risk Assessment ,Article ,Betacoronavirus ,Environmental health ,Occupational Exposure ,Pandemic ,Health care ,London ,Medicine ,Humans ,Prospective Studies ,Seroconversion ,Pandemics ,Cross Infection ,business.industry ,SARS-CoV-2 ,COVID-19 ,General Medicine ,Occupational exposure ,business ,Coronavirus Infections - Published
- 2020
35. Scalable and robust SARS-CoV-2 testing in an academic center
- Author
-
Jim, Aitken, Karen, Ambrose, Sam, Barrell, Rupert, Beale, Ganka, Bineva-Todd, Dhruva, Biswas, Richard, Byrne, Simon, Caidan, Peter, Cherepanov, Laura, Churchward, Graham, Clark, Margaret, Crawford, Laura, Cubitt, Vicky, Dearing, Christopher, Earl, Amelia, Edwards, Chris, Ekin, Efthymios, Fidanis, Alessandra, Gaiba, Steve, Gamblin, Sonia, Gandhi, Jacki, Goldman, Robert, Goldstone, Paul R, Grant, Maria, Greco, Judith, Heaney, Steve, Hindmarsh, Catherine F, Houlihan, Michael, Howell, Michael, Hubank, Deborah, Hughes, Rachael, Instrell, Deborah, Jackson, Mariam, Jamal-Hanjani, Ming, Jiang, Mark, Johnson, Leigh, Jones, Nnennaya, Kanu, George, Kassiotis, Stuart, Kirk, Svend, Kjaer, Andrew, Levett, Lisa, Levett, Marcel, Levi, Wei-Ting, Lu, James I, MacRae, John, Matthews, Laura E, McCoy, Catherine, Moore, David, Moore, Eleni, Nastouli, Jerome, Nicod, Luke, Nightingale, Jessica, Olsen, Nicola, O'Reilly, Amar, Pabari, Venizelos, Papayannopoulos, Namita, Patel, Nigel, Peat, Marc, Pollitt, Peter, Ratcliffe, Caetano, Reis E Sousa, Annachiara, Rosa, Rachel, Rosenthal, Chloe, Roustan, Andrew, Rowan, Gee Yen, Shin, Daniel M, Snell, Ok-Ryul, Song, Moira J, Spyer, Amy, Strange, Charles, Swanton, James M A, Turner, Melanie, Turner, Andreas, Wack, Philip A, Walker, Sophia, Ward, Wai Keong, Wong, Joshua, Wright, Mary, Wu, and Rachel, Zillwood
- Subjects
Europe ,COVID-19 Testing ,Medical research ,Clinical Laboratory Techniques ,Reverse Transcriptase Polymerase Chain Reaction ,Academies and Institutes ,Medical Laboratory Science ,Health care ,Humans ,Coronavirus Infections ,Author Correction ,United Kingdom - Published
- 2020
36. Type I and III interferons disrupt lung epithelial repair during recovery from viral infection
- Author
-
Stefania Crotta, Andreas Wack, Teresa M McCabe, Rune Hartmann, Miriam Llorian, Jack Major, and Hans Henrik Gad
- Subjects
0303 health sciences ,Lung ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Regeneration (biology) ,Endogeny ,Viral infection ,3. Good health ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunopathology ,Immunology ,Medicine ,business ,Beta (finance) ,030304 developmental biology - Abstract
Excessive cytokine signalling frequently exacerbates lung tissue damage during respiratory viral infection. Type I and III interferons (IFN-α/β and IFN-λ) are host-produced antiviral cytokines and currently considered as COVID-19 therapy. Prolonged IFN-α/β responses can lead to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, inducing localised antiviral immunity. Here we show that IFN signalling interferes with lung repair during influenza recovery, with IFN-λ driving these effects most potently. IFN-induced p53 directly reduces epithelial proliferation and differentiation, increasing disease severity and susceptibility to bacterial superinfections. Hence, excessive or prolonged IFN-production aggravates viral infection by impairing lung epithelial regeneration. Therefore, timing and duration are critical parameters of endogenous IFN action, and should be considered carefully for IFN therapeutic strategies against viral infections like influenza and COVID-19.One Sentence SummaryA novel IFN-mediated mechanism of immunopathology during respiratory virus infection by interference with lung tissue repair.
- Published
- 2020
37. Scalable and Resilient SARS-CoV-2 testing in an Academic Centre
- Author
-
Charles Swanton, James I. MacRae, Nnennaya Kanu, Steve Hindmarsh, Karen Ambrose, Andrew Levett, Judith Heaney, Maria Greco, Christopher Earl, Stuart Kirk, Rachel Instrell, Simon Caidan, Peter Cherepanov, Vicky Dearing, Wei-Ting Lu, Laura E. McCoy, George Kassiotis, Mary Wu, Amelia Edwards, Andrew Rowan, Michael Howell, Jessica Olsen, Venizelos Papayannopoulos, Amar Pabari, Caetano Reis e Sousa, Wai Keong Wong, Nicola O’Reilly, Marc Pollitt, Jerome Nicod, Philip A. Walker, Ming Jiang, Marg Crawford, Ganka Bineva-Todd, Alessandra Gaiba, Melanie Turner, Jacki Goldman, James M. A. Turner, Chloe Roustan, Sonia Gandhi, Catherine Moore, Nigel Peat, Rachel Rosenthal, John N. S. Matthews, Amy Strange, Chris Ekin, Marcel Levi, Richard D. Byrne, Laura Churchward, Steve Gamblin, Graham Clark, Jim Aitken, Deb Jackson, Mark Johnson, Svend Kjaer, Joshua Wright, Annachiara Rosa, Andreas Wack, Ok-Ryul Song, Laura Cubitt, Mariam Jamal-Hanjani, David Moore, Leigh Jones, Debbie Hughes, Paul Grant, Sophie Ward, Efthymios Fidanis, Namita Patel, Eleni Nastouli, Dhruva Biswas, Gee Yen Shin, Daniel M Snell, Mike Hubank, Lisa Levett, Moria Spyer, Peter J. Ratcliffe, Luke Nightingale, Sam Barrell, Rupert Beale, Robert L. Goldstone, and Catherine F Houlihan
- Subjects
0303 health sciences ,business.industry ,Computer science ,medicine.disease ,Pipeline (software) ,Turnaround time ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Health care ,Scalability ,Pandemic ,medicine ,Web application ,030212 general & internal medicine ,Medical emergency ,Diagnostic laboratory ,business ,Bespoke ,030304 developmental biology - Abstract
The emergence of the novel coronavirus SARS-CoV-2 has led to a pandemic infecting more than two million people worldwide in less than four months, posing a major threat to healthcare systems. This is compounded by the shortage of available tests causing numerous healthcare workers to unnecessarily self-isolate. We provide a roadmap instructing how a research institute can be repurposed in the midst of this crisis, in collaboration with partner hospitals and an established diagnostic laboratory, harnessing existing expertise in virus handling, robotics, PCR, and data science to derive a rapid, high throughput diagnostic testing pipeline for detecting SARS-CoV-2 in patients with suspected COVID-19. The pipeline is used to detect SARS-CoV-2 from combined nose-throat swabs and endotracheal secretions/ bronchoalveolar lavage fluid. Notably, it relies on a series of in-house buffers for virus inactivation and the extraction of viral RNA, thereby reducing the dependency on commercial suppliers at times of global shortage. We use a commercial RT-PCR assay, from BGI, and results are reported with a bespoke online web application that integrates with the healthcare digital system. This strategy facilitates the remote reporting of thousands of samples a day with a turnaround time of under 24 hours, universally applicable to laboratories worldwide.
- Published
- 2020
38. COVID-19 and emerging viral infections: The case for interferon lambda
- Author
-
Joan E. Durbin, Nancy C. Reich, Andreas Wack, Helen M. Lazear, Jeffrey S. Glenn, Charlotte Odendall, Olusegun O. Onabajo, Sergei V. Kotenko, Deanna M. Santer, Thomas R. O'Brien, Rune Hartmann, Helen Piontkivska, Ludmila Prokunina-Olsson, Ivan Zanoni, Noémie Alphonse, Ruth E Dickenson, Prokunina-Olsson, L, Alphonse, N, Dickenson, R, Durbin, J, Glenn, J, Hartmann, R, Kotenko, S, Lazear, H, O'Brien, T, Odendall, C, Onabajo, O, Piontkivska, H, Santer, D, Reich, N, Wack, A, and Zanoni, I
- Subjects
0301 basic medicine ,antivirus agent ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Pneumonia, Viral ,Immunology ,Virus diseases ,Infectious Disease and Host Defense ,Betacoronavirus ,03 medical and health sciences ,coronavirus disease 2019 ,Viewpoint ,0302 clinical medicine ,Interferon ,Pandemic ,medicine ,Humans ,Immunology and Allergy ,Pandemics ,biology ,SARS-CoV-2 ,business.industry ,COVID-19 ,interferon ,Virus Internalization ,medicine.disease ,biology.organism_classification ,Virology ,3. Good health ,Pneumonia ,030104 developmental biology ,030220 oncology & carcinogenesis ,Interferons ,Coronavirus Infections ,business ,medicine.drug - Abstract
The Viewpoint discusses the benefits and potential limitations of using interferon lambda (IFN-λ, a type III interferon) to prevent, limit, and treat COVID-19. Due to the restricted expression of its receptor complex, IFN-λ provides localized antiviral protection to epithelial cells, including in the respiratory tract., With the first reports on coronavirus disease 2019 (COVID-19), which is caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the scientific community working in the field of type III IFNs (IFN-λ) realized that this class of IFNs could play an important role in this and other emerging viral infections. In this Viewpoint, we present our opinion on the benefits and potential limitations of using IFN-λ to prevent, limit, and treat these dangerous viral infections.
- Published
- 2020
39. Rotavirus susceptibility of antibiotic-treated mice ascribed to diminished expression of interleukin-22
- Author
-
Tanel Mahlakõiv, Stefan T. Peterson, Laure Dumoutier, Ian E Gentle, Peter Staeheli, Celso A. Reis, Meagan E Sullender, Camille Michiels, Pedro P. Hernandez, Annette Ohnemus, Megan T. Baldridge, Stefania Crotta, Andreas Diefenbach, Andreas Wack, Daniel Schnepf, and UCL - SSS/DDUV/MEXP - Médecine expérimentale
- Subjects
Male ,Rotavirus ,Science ,Necroptosis ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Rotavirus Infections ,Microbiology ,Interleukin 22 ,Mice ,Interferon ,medicine ,Animals ,Microbiome ,Intestinal Mucosa ,Mice, Knockout ,Multidisciplinary ,Innate immune system ,Effector ,Gene Expression Profiling ,Interleukins ,Pyroptosis ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Medicine ,Female ,Disease Susceptibility ,Research Article ,medicine.drug - Abstract
The commensal microbiota regulates susceptibility to enteric pathogens by fine-tuning mucosal innate immune responses, but how susceptibility to enteric viruses is shaped by the microbiota remains incompletely understood. Past reports have indicated that commensal bacteria may either promote or repress rotavirus replication in the small intestine of mice. We now report that rotavirus replicated more efficiently in the intestines of germ-free and antibiotic-treated mice compared to animals with an unmodified microbiota. Antibiotic treatment also facilitated rotavirus replication in type I and type III interferon (IFN) receptor-deficient mice, revealing IFN-independent proviral effects. Expression of interleukin-22 (IL-22) was strongly diminished in the intestine of antibiotic-treated mice. Treatment with exogenous IL-22 blocked rotavirus replication in microbiota-depleted wild-type and Stat1-/- mice, demonstrating that the antiviral effect of IL-22 in animals with altered microbiome is not dependent on IFN signaling. In antibiotic-treated animals, IL-22-induced a specific set of genes including Fut2, encoding fucosyl-transferase 2 that participates in the biosynthesis of fucosylated glycans which can mediate rotavirus binding. Interestingly, IL-22 also blocked rotavirus replication in antibiotic-treated Fut2-/- mice. Furthermore, IL-22 inhibited rotavirus replication in antibiotic-treated mice lacking key molecules of the necroptosis or pyroptosis pathways of programmed cell death. Taken together, our results demonstrate that IL-22 determines rotavirus susceptibility of antibiotic-treated mice, yet the IL-22-induced effector molecules conferring rotavirus resistance remain elusive.
- Published
- 2021
40. Monocytes work harder under pressure
- Author
-
Andreas Wack and Pavel Tolar
- Subjects
Work (electrical) ,Chemistry ,Macrophages ,Immunology ,Immunity ,Immunology and Allergy ,Humans ,Data science ,Lung ,Ion Channels ,Monocytes - Published
- 2019
41. Critical requirement for BCR, BAFF, and BAFFR in memory B cell survival
- Author
-
Hamid Mohammadi, Jennifer Müller-Winkler, Richard Mitter, Edina Schweighoffer, Victor L. J. Tybulewicz, Lesley Vanes, Julie C.F. Rappe, and Andreas Wack
- Subjects
Cell Survival ,Immunology ,Naive B cell ,Receptors, Antigen, B-Cell ,Insights ,Article ,Mice ,Antigen ,hemic and lymphatic diseases ,B-Cell Activating Factor ,polycyclic compounds ,Immunology and Allergy ,Animals ,skin and connective tissue diseases ,B-cell activating factor ,Receptor ,Memory B cell ,B-Lymphocytes ,Mice, Inbred BALB C ,biology ,breakpoint cluster region ,bacterial infections and mycoses ,Mice, Inbred C57BL ,biology.protein ,Cancer research ,bacteria ,Antibody ,Signal transduction ,Immunologic Memory ,B-Cell Activation Factor Receptor ,Signal Transduction - Abstract
Memory B cells are long-lived components of humoral immunological memory. Müller-Winkler et al. show that the survival of MBCs is critically dependent on signaling from the BCR, BAFF, BAFFR, and IKK2 kinase., Memory B cells (MBCs) are long-lived cells that form a critical part of immunological memory, providing rapid antibody responses to recurring infections. However, very little is known about signals controlling MBC survival. Previous work has shown that antigen is not required for MBC survival, but a requirement for the B cell antigen receptor (BCR) has not been tested. Other studies have shown that, unlike naive B cells, MBCs do not express BAFFR and their survival is independent of BAFF, the ligand for BAFFR. Here, using inducible genetic ablation, we show that survival of MBCs is critically dependent on the BCR and on signaling through the associated CD79A protein. Unexpectedly, we found that MBCs express BAFFR and that their survival requires BAFF and BAFFR; hence, loss of BAFF or BAFFR impairs recall responses. Finally, we show that MBC survival requires IKK2, a kinase that transduces BAFFR signals. Thus, MBC survival is critically dependent on signaling from BCR and BAFFR.
- Published
- 2019
42. Transcriptional profiling unveils type I and II interferon networks in blood and tissues across diseases
- Author
-
Eleanor Herbert, Damien Chaussabel, Laura Conejero, Katrin D. Mayer-Barber, Alan Sher, Jean Langhorne, Jonathan M. Pitt, Leona Gabryšová, Clare M. Lloyd, Jacques Banchereau, Simon L. Priestnall, Virginia Pascual, Qian Wang, Venizelos Papayannopoulos, Evangelos Stavropoulos, Jonathan S. Dodd, Lúcia Moreira-Teixeira, Sophia Davidson, Eva M. Frickel, Dragana Jankovic, Annika Warnatsch, Jing-wen Lin, Probir Chakravarty, Akul Singhania, William J. Branchett, Andreas Wack, Nicole Baldwin, Marianna Ioannou, Ian R. Humphreys, Christine M. Graham, Peter J. M. Openshaw, Anne O'Garra, Gregory J. Bancroft, Mark S. Wilson, and Abdul Sesay
- Subjects
0301 basic medicine ,Burkholderia pseudomallei ,HOST-RESISTANCE ,General Physics and Astronomy ,02 engineering and technology ,Receptor, Interferon alpha-beta ,Pathogenesis ,Mice ,Interferon ,Gene expression ,Candida albicans ,IMMUNE-RESPONSE ,lcsh:Science ,Lung ,NEUTROPHILS ,Receptors, Interferon ,Chemical Biology & High Throughput ,Human Biology & Physiology ,IFN-GAMMA ,Multidisciplinary ,Effector ,INDUCTION ,Genome Integrity & Repair ,Candidiasis ,respiratory system ,021001 nanoscience & nanotechnology ,Multidisciplinary Sciences ,medicine.anatomical_structure ,Interferon Type I ,Science & Technology - Other Topics ,Infectious diseases ,medicine.symptom ,0210 nano-technology ,Genetics & Genomics ,medicine.drug ,Model organisms ,Science ,Immunology ,Inflammation ,chemical and pharmacologic phenomena ,Infectious Disease ,Respiratory Syncytial Virus Infections ,Granulocyte ,Biology ,TUBERCULOSIS ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Interferon-gamma ,Immune system ,Orthomyxoviridae Infections ,MD Multidisciplinary ,medicine ,Animals ,Gene ,Computational & Systems Biology ,Science & Technology ,Influenza A Virus, H3N2 Subtype ,FOS: Clinical medicine ,LUNG INFLAMMATION ,General Chemistry ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Tumour Biology ,Computational biology and bioinformatics ,Mice, Inbred C57BL ,Gene regulation in immune cells ,030104 developmental biology ,TOXOPLASMA-GONDII ,Gene Expression Regulation ,Melioidosis ,CELLS ,lcsh:Q - Abstract
Understanding how immune challenges elicit different responses is critical for diagnosing and deciphering immune regulation. Using a modular strategy to interpret the complex transcriptional host response in mouse models of infection and inflammation, we show a breadth of immune responses in the lung. Lung immune signatures are dominated by either IFN-γ and IFN-inducible, IL-17-induced neutrophil- or allergy-associated gene expression. Type I IFN and IFN-γ-inducible, but not IL-17- or allergy-associated signatures, are preserved in the blood. While IL-17-associated genes identified in lung are detected in blood, the allergy signature is only detectable in blood CD4+ effector cells. Type I IFN-inducible genes are abrogated in the absence of IFN-γ signaling and decrease in the absence of IFNAR signaling, both independently contributing to the regulation of granulocyte responses and pathology during Toxoplasma gondii infection. Our framework provides an ideal tool for comparative analyses of transcriptional signatures contributing to protection or pathogenesis in disease., The authors present an extensive profile of host transcriptional respones to a diverse group of pathogens and allergens. In doing so, they identify TH1, type I IFN, TH17, and TH2 responses, that underlie each immune response in both the blood and lung, which represents a global profile of host-pathogen immune responses.
- Published
- 2019
43. Influenza-induced monocyte-derived alveolar macrophages confer prolonged antibacterial protection
- Author
-
Matthias Mack, Stefania Crotta, Andreas Wack, Justina Kulikauskaite, Soren Beinke, Edith M. Hessel, Harshil Patel, Gavin Kelly, and Helena Aegerter
- Subjects
0301 basic medicine ,Immunology ,Population ,medicine.disease_cause ,Pneumococcal Infections ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Orthomyxoviridae Infections ,Immunity ,Streptococcus pneumoniae ,Macrophages, Alveolar ,Immunology and Allergy ,Medicine ,Animals ,Respiratory system ,education ,education.field_of_study ,Lung ,business.industry ,Monocyte derived ,Phenotype ,Immunity, Innate ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,business ,030215 immunology - Abstract
Despite the prevalence and clinical importance of influenza, its long-term effect on lung immunity is unclear. Here we describe that following viral clearance and clinical recovery, at 1 month after infection with influenza, mice are better protected from Streptococcus pneumoniae infection due to a population of monocyte-derived alveolar macrophages (AMs) that produce increased interleukin-6. Influenza-induced monocyte-derived AMs have a surface phenotype similar to resident AMs but display a unique functional, transcriptional and epigenetic profile that is distinct from resident AMs. In contrast, influenza-experienced resident AMs remain largely similar to naive AMs. Thus, influenza changes the composition of the AM population to provide prolonged antibacterial protection. Monocyte-derived AMs persist over time but lose their protective profile. Our results help to understand how transient respiratory infections, a common occurrence in human life, can constantly alter lung immunity by contributing monocyte-derived, recruited cells to the AM population. Respiratory infections occur throughout life but how this shapes the lung immune system through time is unclear. Wack and colleagues show that a previous influenza infection recruits monocytes to the lung, which then assume an alveolar macrophage-like phenotype and mediate long-term antibacterial protection.
- Published
- 2018
44. Multiple levels of control determine how E4bp4/Nfil3 regulates NK cell development
- Author
-
Andreas Wack, Peter A. DiMaggio, Tomasz Kostrzewski, Aaron J. Borg, Iva Filipovic, Yiran Meng, Victoria Male, and Hugh J.M. Brady
- Subjects
0301 basic medicine ,Immunology ,Cell ,Notch signaling pathway ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,Receptor, Notch1 ,Transcription factor ,Mice, Knockout ,Cell growth ,Innate lymphoid cell ,NFIL3 ,HEK 293 cells ,Cell Differentiation ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Basic-Leucine Zipper Transcription Factors ,HEK293 Cells ,Gene Expression Regulation ,Protein Processing, Post-Translational ,030215 immunology ,HeLa Cells - Abstract
The transcription factor E4bp4/Nfil3 has been shown to have a critical role in the development of all innate lymphoid cell types including NK cells. In this study, we show that posttranslational modifications of E4bp4 by either SUMOylation or phosphorylation have profound effects on both E4bp4 function and NK cell development. We examined the activity of E4bp4 mutants lacking posttranslational modifications and found that Notch1 was a novel E4bp4 target gene. We observed that abrogation of Notch signaling impeded NK cell production and the total lack of NK cell development from E4bp4−/− progenitors was completely rescued by short exposure to Notch peptide ligands. This work reveals both novel mechanisms in NK cell development by a transcriptional network including E4bp4 with Notch, and that E4bp4 is a central hub to process extrinsic stimuli.
- Published
- 2018
45. TRAIL + monocytes and monocyte‐related cells cause lung damage and thereby increase susceptibility to influenza– <scp>S</scp> treptococcus pneumoniae coinfection
- Author
-
Stefania Crotta, Venizelos Papayannopoulos, Gregory T Ellis, Sophia Davidson, Andreas Wack, and Nora Branzk
- Subjects
0303 health sciences ,Monocyte ,Biology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,3. Good health ,Proinflammatory cytokine ,Pathogenesis ,Epithelial Damage ,03 medical and health sciences ,Pneumococcal infections ,0302 clinical medicine ,medicine.anatomical_structure ,Streptococcus pneumoniae ,Immunology ,Genetics ,medicine ,Coinfection ,Tumor necrosis factor alpha ,Molecular Biology ,030304 developmental biology ,030215 immunology - Abstract
Streptococcus pneumoniae coinfection is a major cause of influenza-associated mortality; however, the mechanisms underlying pathogenesis or protection remain unclear. Using a clinically relevant mouse model, we identify immune-mediated damage early during coinfection as a new mechanism causing susceptibility. Coinfected CCR2−/− mice lacking monocytes and monocyte-derived cells control bacterial invasion better, show reduced epithelial damage and are overall more resistant than wild-type controls. In influenza-infected wild-type lungs, monocytes and monocyte-derived cells are the major cell populations expressing the apoptosis-inducing ligand TRAIL. Accordingly, anti-TRAIL treatment reduces bacterial load and protects against coinfection if administered during viral infection, but not following bacterial exposure. Post-influenza bacterial outgrowth induces a strong proinflammatory cytokine response and massive inflammatory cell infiltrate. Depletion of neutrophils or blockade of TNF-α facilitate bacterial outgrowth, leading to increased mortality, demonstrating that these factors aid bacterial control. We conclude that inflammatory monocytes recruited early, during the viral phase of coinfection, induce TRAIL-mediated lung damage, which facilitates bacterial invasion, while TNF-α and neutrophil responses help control subsequent bacterial outgrowth. We thus identify novel determinants of protection versus pathology in influenza–Streptococcus pneumoniae coinfection.
- Published
- 2015
46. Guarding the frontiers: the biology of type III interferons
- Author
-
Andreas Wack, Ewa Terczyńska-Dyla, and Rune Hartmann
- Subjects
Innate immunity ,Inflammation ,Innate immune system ,Human liver ,Immune protection ,Interleukins ,Hepatitis C virus ,Immunology ,Models, Immunological ,Hepacivirus ,Inflammatory diseases ,Biology ,medicine.disease_cause ,Hepatitis C ,Article ,Hepatocytes ,medicine ,Humans ,Immunology and Allergy ,Interferons ,medicine.symptom ,Chromosomes, Human, Pair 19 ,Gene - Abstract
Type I and III interferons share similar antiviral properties, but there are some important distinctions. Hartmann and colleagues review the specialized functions of type III interferons, including their ability to mediate antiviral functions at barrier surfaces., Type III interferons (IFNs) or IFN-λs regulate a similar set of genes as type I IFNs, but whereas type I IFNs act globally, IFN-λs primarily target mucosal epithelial cells and protect them against the frequent viral attacks that are typical for barrier tissues. IFN-λs thereby help to maintain healthy mucosal surfaces through immune protection, without the significant immune-related pathogenic risk associated with type I IFN responses. Type III IFNs also target the human liver, with dual effects: they induce an antiviral state in hepatocytes, but specific IFN-λ4 action impairs the clearance of hepatitis C virus and could influence inflammatory responses. This constitutes a paradox that has yet to be resolved.
- Published
- 2015
47. A Serpin Shapes the Extracellular Environment to Prevent Influenza A Virus Maturation
- Author
-
Charles M. Rice, Michael J. Ciancanelli, Jing W. Xiao, Kierstin L. Bell, Hans-Heinrich Hoffmann, Yingpu Yu, Jean-Laurent Casanova, Brenna Flatley, Paul D. Bieniasz, Sam J. Wilson, Meike Dittmann, Stefania Crotta, Margaret A. Scull, Andreas Wack, and Rachel H. Gilmore
- Subjects
Proteases ,Respiratory System ,Serpin ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,chemistry.chemical_compound ,Viral life cycle ,Plasminogen Activator Inhibitor 1 ,Serpin E2 ,Extracellular ,Influenza A virus ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,Biochemistry, Genetics and Molecular Biology(all) ,Effector ,Virology ,Immunity, Innate ,3. Good health ,Mice, Inbred C57BL ,chemistry ,Plasminogen activator inhibitor-1 ,Serine Proteases ,Glycoprotein - Abstract
SummaryInterferon-stimulated genes (ISGs) act in concert to provide a tight barrier against viruses. Recent studies have shed light on the contribution of individual ISG effectors to the antiviral state, but most have examined those acting on early, intracellular stages of the viral life cycle. Here, we applied an image-based screen to identify ISGs inhibiting late stages of influenza A virus (IAV) infection. We unraveled a directly antiviral function for the gene SERPINE1, encoding plasminogen activator inhibitor 1 (PAI-1). By targeting extracellular airway proteases, PAI-1 inhibits IAV glycoprotein cleavage, thereby reducing infectivity of progeny viruses. This was biologically relevant for IAV restriction in vivo. Further, partial PAI-1 deficiency, attributable to a polymorphism in human SERPINE1, conferred increased susceptibility to IAV in vitro. Together, our findings reveal that manipulating the extracellular environment to inhibit the last step in a virus life cycle is an important mechanism of the antiviral response.
- Published
- 2015
48. Type I interferons in infectious disease
- Author
-
Finlay W. McNab, Katrin D. Mayer-Barber, Anne O'Garra, Alan Sher, and Andreas Wack
- Subjects
History ,Tuberculosis ,medicine.medical_treatment ,Biology ,Communicable Diseases ,Article ,Virus ,Education ,Mycobacterium tuberculosis ,Immune system ,Immunopathology ,Parasitic Diseases ,medicine ,Animals ,Humans ,B cell ,Immunosuppression ,Bacterial Infections ,medicine.disease ,biology.organism_classification ,Virology ,Computer Science Applications ,medicine.anatomical_structure ,Mycoses ,Virus Diseases ,Infectious disease (medical specialty) ,Interferon Type I ,Immunology ,Signal Transduction - Abstract
Most, if not all, cells in humans and mice express the receptor for type I interferons (IFNs). Therefore, these cytokines have a range of direct and indirect effects on various cell types during infection with viruses, bacteria, parasites and fungi. Type I IFNs are important for host defence against viruses, through the induction of antiviral effector molecules that are encoded by IFN-stimulated genes. These IFNs can, however, cause immunopathology in acute viral infections. Conversely, they can lead to immunosuppression and loss of virus control during chronic viral infections. During bacterial infections, low levels of type I IFNs may be required early, to initiate cell-mediated immune responses. By contrast, type I IFNs have been shown to have adverse effects in infections with intracellular bacteria such as Listeria monocytogenes and Mycobacterium tuberculosis. In bacterial infections, high concentrations of type I IFNs may block B cell responses or may lead to the production of immunosuppressive molecules such as interleukin-10. Type I IFNs also antagonize the action of type II IFN (that is, IFNγ) by reducing the responsiveness of macrophages to activation by type II IFN. Another important antagonism is between type I IFNs and interleukin-1. This antagonism was recently shown to be important in M. tuberculosis infection and to be mediated by eicosanoids, in particular prostaglandin E2. Thus, type I IFNs are part of a complex cross-regulatory network, which leads mostly, but not always, to protection of the host against infectious diseases with minimum damage to the host.
- Published
- 2015
49. Disease-Promoting Effects of Type I Interferons in Viral, Bacterial, and Coinfections
- Author
-
Andreas Wack, Sophia Davidson, and Mala K. Maini
- Subjects
Programmed cell death ,Effector ,Coinfection ,medicine.medical_treatment ,Immunology ,Reviews ,Inflammation ,Immunosuppression ,Cell Biology ,Disease ,Bacterial Infections ,Biology ,3. Good health ,Immune system ,Virus Diseases ,Virology ,Immunopathology ,Interferon Type I ,medicine ,Humans ,medicine.symptom ,Interferon type I ,medicine.drug - Abstract
While type I interferons (IFNs) are universally acknowledged for their antiviral and immunostimulatory functions, there is increasing appreciation of the detrimental effects of inappropriate, excessive, or mistimed type I IFN responses in viral and bacterial infections. The underlying mechanisms by which type I IFNs promote susceptibility or severity include direct tissue damage by apoptosis induction or suppression of proliferation in tissue cells, immunopathology due to excessive inflammation, and cell death induced by TRAIL- and Fas-expressing immune cells, as well as immunosuppression through IL-10, IL-27, PD-L1, IL-1Ra, and other regulatory molecules that antagonize the induction or action of IL-1, IL-12, IL-17, IFN-γ, KC, and other effectors of the immune response. Bacterial superinfections following influenza infection are a prominent example of a situation where type I IFNs can misdirect the immune response. This review discusses current understanding of the parameters of signal strength, duration, timing, location, and cellular recipients that determine whether type I IFNs have beneficial or detrimental effects in infection.
- Published
- 2015
50. The Transcription Factor E4BP4 Is Not Required for Extramedullary Pathways of NK Cell Development
- Author
-
Sophia Davidson, Victoria Male, Hugh J.M. Brady, Andreas Wack, Stefania Crotta, João H. Duarte, Annita Gkioka, Ilaria Nisoli, and Medical Research Council (MRC)
- Subjects
Liver cytology ,T-Lymphocytes ,Cellular differentiation ,Immunology ,INNATE LYMPHOID-CELLS ,Eomesodermin ,Thymus Gland ,Biology ,MURINE BONE-MARROW ,T-CELL ,MATURATION ,Cell Line ,ACTIVATION ,Interleukin 21 ,Animals ,Immunology and Allergy ,Progenitor cell ,UNIQUE SUBSET ,Cells, Cultured ,NATURAL-KILLER-CELL ,Homeodomain Proteins ,Mice, Knockout ,Science & Technology ,IDENTIFICATION ,PROGENITORS ,Interleukin-7 ,Janus kinase 3 ,Immune System Development ,Cell Differentiation ,Flow Cytometry ,3. Good health ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,MICE ,Thymocyte ,Basic-Leucine Zipper Transcription Factors ,Liver ,1107 Immunology ,Interleukin 12 ,T-Box Domain Proteins ,Life Sciences & Biomedicine ,Spleen ,Signal Transduction - Abstract
NK cells contribute to antitumor and antiviral immunosurveillance. Their development in the bone marrow (BM) requires the transcription factor E4BP4/NFIL3, but requirements in other organs are less well defined. In this study, we show that CD3−NK1.1+NKp46+CD122+ NK cells of immature phenotype and expressing low eomesodermin levels are found in thymus, spleen, and liver of E4BP4-deficient mice, whereas numbers of mature, eomesoderminhigh conventional NK cells are drastically reduced. E4BP4-deficient CD44+CD25− double-negative 1 thymocytes efficiently develop in vitro into NK cells with kinetics, phenotype, and functionality similar to wild-type controls, whereas no NK cells develop from E4BP4-deficient BM precursors. In E4BP4/Rag-1 double-deficient (DKO) mice, NK cells resembling those in Rag-1–deficient controls are found in similar numbers in the thymus and liver. However, NK precursors are reduced in DKO BM, and no NK cells develop from DKO BM progenitors in vitro. DKO thymocyte precursors readily develop into NK cells, but DKO BM transfers into nude recipients and NK cells in E4BP4/Rag-1/IL-7 triple-KO mice indicated thymus-independent NK cell development. In the presence of T cells or E4BP4-sufficient NK cells, DKO NK cells have a selective disadvantage, and thymic and hepatic DKO NK cells show reduced survival when adoptively transferred into lymphopenic hosts. This correlates with higher apoptosis rates and lower responsiveness to IL-15 in vitro. In conclusion, we demonstrate E4BP4-independent development of NK cells of immature phenotype, reduced fitness, short t1/2, and potential extramedullary origin. Our data identify E4BP4-independent NK cell developmental pathways and a role for E4BP4 in NK cell homeostasis.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.