LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 28

Search options

  1. Article ; Online: Virus-induced metabolic reprogramming and innate sensing hereof by the infected host.

    Thyrsted, Jacob / Holm, Christian Kanstrup

    Current opinion in biotechnology

    2020  Volume 68, Page(s) 44–50

    Abstract: To make new infectious particles, all viruses must manipulate host cell metabolism to secure sufficient availability of biomolecules and energy-a phenomenon now known as metabolic reprogramming. Numerous observations of this has already been made for a ... ...

    Abstract To make new infectious particles, all viruses must manipulate host cell metabolism to secure sufficient availability of biomolecules and energy-a phenomenon now known as metabolic reprogramming. Numerous observations of this has already been made for a range of viruses with each type of virus seemingly applying its own unique tactics to accomplish this unifying goal. In this light, metabolic reprogramming of the infected cell is largely beneficial to the virus and not to the host. On the other hand, virus-induced metabolic reprogramming represents a transformed self with distorted cellular and extracellular levels of distinct metabolites and metabolic by-products. This review briefly outlines current knowledge of virus-induced metabolic reprogramming, discusses how this could be sensed by the infected host to initiate anti-viral programs, and presents examples of innate anti-viral mechanisms of the host that target the availability of biomolecules to block viral replication.
    MeSH term(s) Host-Pathogen Interactions ; Immunity, Innate ; Virus Replication ; Viruses
    Language English
    Publishing date 2020-10-25
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1052045-4
    ISSN 1879-0429 ; 0958-1669
    ISSN (online) 1879-0429
    ISSN 0958-1669
    DOI 10.1016/j.copbio.2020.10.004
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: NRF2 in Viral Infection.

    Herengt, Angela / Thyrsted, Jacob / Holm, Christian K

    Antioxidants (Basel, Switzerland)

    2021  Volume 10, Issue 9

    Abstract: The transcription factor NRF2 is central to redox homeostasis in animal cells and is a well-known driver of chemoresistance in many types of cancer. Recently, new roles have been ascribed to NRF2 which include regulation of antiviral interferon responses ...

    Abstract The transcription factor NRF2 is central to redox homeostasis in animal cells and is a well-known driver of chemoresistance in many types of cancer. Recently, new roles have been ascribed to NRF2 which include regulation of antiviral interferon responses and inflammation. In addition, NRF2 is emerging as an important factor in antiviral immunity through interferon-independent mechanisms. In the review, we give an overview of the scientific progress on the involvement and importance of NRF2 in the context of viral infection.
    Language English
    Publishing date 2021-09-18
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2704216-9
    ISSN 2076-3921
    ISSN 2076-3921
    DOI 10.3390/antiox10091491
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: NRF2 in Viral Infection

    Herengt, Angela / Thyrsted, Jacob / Holm, Christian K.

    Antioxidants. 2021 Sept. 18, v. 10, no. 9

    2021  

    Abstract: The transcription factor NRF2 is central to redox homeostasis in animal cells and is a well-known driver of chemoresistance in many types of cancer. Recently, new roles have been ascribed to NRF2 which include regulation of antiviral interferon responses ...

    Abstract The transcription factor NRF2 is central to redox homeostasis in animal cells and is a well-known driver of chemoresistance in many types of cancer. Recently, new roles have been ascribed to NRF2 which include regulation of antiviral interferon responses and inflammation. In addition, NRF2 is emerging as an important factor in antiviral immunity through interferon-independent mechanisms. In the review, we give an overview of the scientific progress on the involvement and importance of NRF2 in the context of viral infection.
    Keywords homeostasis ; immunity ; inflammation ; interferons ; transcription factors
    Language English
    Dates of publication 2021-0918
    Publishing place Multidisciplinary Digital Publishing Institute
    Document type Article
    ZDB-ID 2704216-9
    ISSN 2076-3921
    ISSN 2076-3921
    DOI 10.3390/antiox10091491
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  4. Article ; Online: Amyloid-β aggregates activate peripheral monocytes in mild cognitive impairment.

    Juul-Madsen, Kristian / Parbo, Peter / Ismail, Rola / Ovesen, Peter L / Schmidt, Vanessa / Madsen, Lasse S / Thyrsted, Jacob / Gierl, Sarah / Breum, Mihaela / Larsen, Agnete / Andersen, Morten N / Romero-Ramos, Marina / Holm, Christian K / Andersen, Gregers R / Zhao, Huaying / Schuck, Peter / Nygaard, Jens V / Sutherland, Duncan S / Eskildsen, Simon F /
    Willnow, Thomas E / Brooks, David J / Vorup-Jensen, Thomas

    Nature communications

    2024  Volume 15, Issue 1, Page(s) 1224

    Abstract: The peripheral immune system is important in neurodegenerative diseases, both in protecting and inflaming the brain, but the underlying mechanisms remain elusive. Alzheimer's Disease is commonly preceded by a prodromal period. Here, we report the ... ...

    Abstract The peripheral immune system is important in neurodegenerative diseases, both in protecting and inflaming the brain, but the underlying mechanisms remain elusive. Alzheimer's Disease is commonly preceded by a prodromal period. Here, we report the presence of large Aβ aggregates in plasma from patients with mild cognitive impairment (n = 38). The aggregates are associated with low level Alzheimer's Disease-like brain pathology as observed by
    MeSH term(s) Humans ; Alzheimer Disease/pathology ; Amyloid beta-Peptides ; Cognitive Dysfunction ; Integrin alphaXbeta2 ; Monocytes/pathology
    Chemical Substances Amyloid beta-Peptides ; Integrin alphaXbeta2
    Language English
    Publishing date 2024-02-09
    Publishing country England
    Document type Journal Article
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-024-45627-y
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Influenza A induces lactate formation to inhibit type I IFN in primary human airway epithelium.

    Thyrsted, Jacob / Storgaard, Jacob / Blay-Cadanet, Julia / Heinz, Alexander / Thielke, Anne Laugaard / Crotta, Stefania / de Paoli, Frank / Olagnier, David / Wack, Andreas / Hiller, Karsten / Hansen, Anne Louise / Holm, Christian Kanstrup

    iScience

    2021  Volume 24, Issue 11, Page(s) 103300

    Abstract: 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 ... ...

    Abstract 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.
    Language English
    Publishing date 2021-10-15
    Publishing country United States
    Document type Journal Article
    ISSN 2589-0042
    ISSN (online) 2589-0042
    DOI 10.1016/j.isci.2021.103300
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Influenza A induces lactate formation to inhibit type I IFN in primary human airway epithelium

    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 / Christian Kanstrup Holm

    iScience, Vol 24, Iss 11, Pp 103300- (2021)

    2021  

    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 ... ...

    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.
    Keywords Immune response ; Virology ; Metabolomics ; Science ; Q
    Subject code 570
    Language English
    Publishing date 2021-11-01T00:00:00Z
    Publisher Elsevier
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  7. Article ; Online: Ionophore antibiotic X-206 is a potent inhibitor of SARS-CoV-2 infection in vitro.

    Svenningsen, Esben B / Thyrsted, Jacob / Blay-Cadanet, Julia / Liu, Han / Lin, Shaoquan / Moyano-Villameriel, Jaime / Olagnier, David / Idorn, Manja / Paludan, Søren R / Holm, Christian K / Poulsen, Thomas B

    Antiviral research

    2020  Volume 185, Page(s) 104988

    Abstract: Pandemic spread of emerging human pathogenic viruses, such as the current SARS-CoV-2, poses both an immediate and future challenge to human health and society. Currently, effective treatment of infection with SARS-CoV-2 is limited and broad spectrum ... ...

    Abstract Pandemic spread of emerging human pathogenic viruses, such as the current SARS-CoV-2, poses both an immediate and future challenge to human health and society. Currently, effective treatment of infection with SARS-CoV-2 is limited and broad spectrum antiviral therapies to meet other emerging pandemics are absent leaving the World population largely unprotected. Here, we have identified distinct members of the family of polyether ionophore antibiotics with potent ability to inhibit SARS-CoV-2 replication and cytopathogenicity in cells. Several compounds from this class displayed more than 100-fold selectivity between viral-induced cytopathogenicity and inhibition of cell viability, however the compound X-206 displayed >500-fold selectivity and was furthermore able to inhibit viral replication even at sub-nM levels. The antiviral mechanism of the polyether ionophores is currently not understood in detail. We demonstrate, e.g. through unbiased bioactivity profiling, that their effects on the host cells differ from those of cationic amphiphiles such as hydroxychloroquine. Collectively, our data suggest that polyether ionophore antibiotics should be subject to further investigations as potential broad-spectrum antiviral agents.
    MeSH term(s) Animals ; Anti-Bacterial Agents/pharmacology ; Antiviral Agents/pharmacology ; COVID-19/drug therapy ; Chlorocebus aethiops ; Ethers, Cyclic/pharmacology ; Humans ; Ionophores/pharmacology ; SARS-CoV-2/drug effects ; Vero Cells ; Virus Replication/drug effects
    Chemical Substances Anti-Bacterial Agents ; Antiviral Agents ; Ethers, Cyclic ; Ionophores ; X-206, polyether antibiotic
    Language English
    Publishing date 2020-11-25
    Publishing country Netherlands
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 306628-9
    ISSN 1872-9096 ; 0166-3542
    ISSN (online) 1872-9096
    ISSN 0166-3542
    DOI 10.1016/j.antiviral.2020.104988
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: SARS-CoV-2 evades immune detection in alveolar macrophages.

    Dalskov, Louise / Møhlenberg, Michelle / Thyrsted, Jacob / Blay-Cadanet, Julia / Poulsen, Ebbe Toftgaard / Folkersen, Birgitte Holst / Skaarup, Søren Helbo / Olagnier, David / Reinert, Line / Enghild, Jan Johannes / Hoffmann, Hans Jürgen / Holm, Christian Kanstrup / Hartmann, Rune

    EMBO reports

    2020  Volume 21, Issue 12, Page(s) e51252

    Abstract: Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune ... ...

    Abstract Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune response to respiratory viruses. AMs are likely the first immune cells to encounter SARS-CoV-2 during an infection, and their reaction to the virus will have a profound impact on the outcome of the infection. Interferons (IFNs) are antiviral cytokines and among the first cytokines produced upon viral infection. In this study, AMs from non-infectious donors are challenged with SARS-CoV-2. We demonstrate that challenged AMs are incapable of sensing SARS-CoV-2 and of producing an IFN response in contrast to other respiratory viruses, like influenza A virus and Sendai virus, which trigger a robust IFN response. The absence of IFN production in AMs upon challenge with SARS-CoV-2 could explain the initial asymptotic phase observed during COVID-19 and argues against AMs being the sources of pro-inflammatory cytokines later during infection.
    MeSH term(s) Antiviral Agents/immunology ; COVID-19/immunology ; COVID-19/virology ; Cells, Cultured ; Cytokines/immunology ; Epithelial Cells/immunology ; Epithelial Cells/virology ; Humans ; Immune Evasion ; Interferon Type I/immunology ; Lung/immunology ; Lung/virology ; Macrophages, Alveolar/immunology ; Macrophages, Alveolar/virology ; Pandemics ; SARS-CoV-2/immunology
    Chemical Substances Antiviral Agents ; Cytokines ; Interferon Type I
    Keywords covid19
    Language English
    Publishing date 2020-10-28
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2020896-0
    ISSN 1469-3178 ; 1469-221X
    ISSN (online) 1469-3178
    ISSN 1469-221X
    DOI 10.15252/embr.202051252
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: TLR2 and TLR7 mediate distinct immunopathological and antiviral plasmacytoid dendritic cell responses to SARS-CoV-2 infection.

    van der Sluis, Renée M / Cham, Lamin B / Gris-Oliver, Albert / Gammelgaard, Kristine R / Pedersen, Jesper G / Idorn, Manja / Ahmadov, Ulvi / Hernandez, Sabina Sanches / Cémalovic, Ena / Godsk, Stine H / Thyrsted, Jacob / Gunst, Jesper D / Nielsen, Silke D / Jørgensen, Janni J / Bjerg, Tobias Wang / Laustsen, Anders / Reinert, Line S / Olagnier, David / Bak, Rasmus O /
    Kjolby, Mads / Holm, Christian K / Tolstrup, Martin / Paludan, Søren R / Kristensen, Lasse S / Søgaard, Ole S / Jakobsen, Martin R

    The EMBO journal

    2022  Volume 41, Issue 10, Page(s) e109622

    Abstract: Understanding the molecular pathways driving the acute antiviral and inflammatory response to SARS-CoV-2 infection is critical for developing treatments for severe COVID-19. Here, we find decreasing number of circulating plasmacytoid dendritic cells ( ... ...

    Abstract Understanding the molecular pathways driving the acute antiviral and inflammatory response to SARS-CoV-2 infection is critical for developing treatments for severe COVID-19. Here, we find decreasing number of circulating plasmacytoid dendritic cells (pDCs) in COVID-19 patients early after symptom onset, correlating with disease severity. pDC depletion is transient and coincides with decreased expression of antiviral type I IFNα and of systemic inflammatory cytokines CXCL10 and IL-6. Using an in vitro stem cell-based human pDC model, we further demonstrate that pDCs, while not supporting SARS-CoV-2 replication, directly sense the virus and in response produce multiple antiviral (interferons: IFNα and IFNλ1) and inflammatory (IL-6, IL-8, CXCL10) cytokines that protect epithelial cells from de novo SARS-CoV-2 infection. Via targeted deletion of virus-recognition innate immune pathways, we identify TLR7-MyD88 signaling as crucial for production of antiviral interferons (IFNs), whereas Toll-like receptor (TLR)2 is responsible for the inflammatory IL-6 response. We further show that SARS-CoV-2 engages the receptor neuropilin-1 on pDCs to selectively mitigate the antiviral interferon response, but not the IL-6 response, suggesting neuropilin-1 as potential therapeutic target for stimulation of TLR7-mediated antiviral protection.
    MeSH term(s) COVID-19/immunology ; COVID-19/pathology ; Cytokines/metabolism ; Dendritic Cells/immunology ; Dendritic Cells/pathology ; Humans ; Interferon Type I/immunology ; Interferon-alpha/immunology ; Interleukin-6/immunology ; Neuropilin-1/immunology ; SARS-CoV-2 ; Toll-Like Receptor 2/immunology ; Toll-Like Receptor 7/immunology
    Chemical Substances Cytokines ; Interferon Type I ; Interferon-alpha ; Interleukin-6 ; TLR2 protein, human ; TLR7 protein, human ; Toll-Like Receptor 2 ; Toll-Like Receptor 7 ; Neuropilin-1 (144713-63-3)
    Language English
    Publishing date 2022-03-01
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 586044-1
    ISSN 1460-2075 ; 0261-4189
    ISSN (online) 1460-2075
    ISSN 0261-4189
    DOI 10.15252/embj.2021109622
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: SARS-CoV-2 evades immune detection in alveolar macrophages

    Dalskov, Louise / Møhlenberg, Michelle / Thyrsted, Jacob / Blay-Cadanet, Julia / Poulsen, Ebbe Toftgaard / Folkersen, Birgitte Holst / Skaarup, Søren Helbo / Olagnier, David / Reinert, Line / Enghild, Jan Johannes / Hoffmann, Hans Jürgen / Holm, Christian Kanstrup / Hartmann, Rune

    EMBO Rep

    Abstract: Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune ... ...

    Abstract Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune response to respiratory viruses. AMs are likely the first immune cells to encounter SARS-CoV-2 during an infection, and their reaction to the virus will have a profound impact on the outcome of the infection. Interferons (IFNs) are antiviral cytokines and among the first cytokines produced upon viral infection. In this study, AMs from non-infectious donors are challenged with SARS-CoV-2. We demonstrate that challenged AMs are incapable of sensing SARS-CoV-2 and of producing an IFN response in contrast to other respiratory viruses, like influenza A virus and Sendai virus, which trigger a robust IFN response. The absence of IFN production in AMs upon challenge with SARS-CoV-2 could explain the initial asymptotic phase observed during COVID-19 and argues against AMs being the sources of pro-inflammatory cytokines later during infection.
    Keywords covid19
    Publisher WHO
    Document type Article
    Note WHO #Covidence: #895751
    Database COVID19

    Kategorien

To top