Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anders Laustsen is active.

Publication


Featured researches published by Anders Laustsen.


Proceedings of the National Academy of Sciences of the United States of America | 2013

IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication

Martin R. Jakobsen; Rasmus O. Bak; Annika Andersen; Randi K. Berg; Søren B. Jensen; Tengchuan Jin; Anders Laustsen; Kathrine Hansen; Lars Østergaard; Katherine A. Fitzgerald; T. Sam Xiao; Jacob Giehm Mikkelsen; Trine H. Mogensen; Søren R. Paludan

Significance HIV-1 is a lentivirus and replicates through a replication cycle involving several DNA forms including ssDNA. Here we report that synthetic DNA oligos corresponding to DNA forms of the lentivirus replication cycle as well as viral DNA are detected by the immunological DNA sensor IFN-inducible protein 16 (IFI16) and stimulate innate immune responses through a pathway dependent on stimulator of IFN genes (STING). Moreover, we show that replication of HIV-1 is elevated in cells with decreased expression of IFI16 or STING. We suggest IFI16 is a sensor for lentivirus DNA in macrophages stimulating innate immune responses, which contribute to early control of the virus. Replication of lentiviruses generates different DNA forms, including RNA:DNA hybrids, ssDNA, and dsDNA. Nucleic acids stimulate innate immune responses, and pattern recognition receptors detecting dsDNA have been identified. However, sensors for ssDNA have not been reported, and the ability of RNA:DNA hybrids to stimulate innate immune responses is controversial. Using ssDNAs derived from HIV-1 proviral DNA, we report that this DNA form potently induces the expression of IFNs in primary human macrophages. This response was stimulated by stem regions in the DNA structure and was dependent on IFN-inducible protein 16 (IFI16), which bound immunostimulatory DNA directly and activated the stimulator of IFN genes –TANK-binding kinase 1 - IFN regulatory factors 3/7 (STING–TBK1–IRF3/7) pathway. Importantly, IFI16 colocalized and associated with lentiviral DNA in the cytoplasm in macrophages, and IFI16 knockdown in this cell type augmented lentiviral transduction and also HIV-1 replication. Thus, IFI16 is a sensor for DNA forms produced during the lentiviral replication cycle and regulates HIV-1 replication in macrophages.


The EMBO Journal | 2014

Listeria monocytogenes induces IFNβ expression through an IFI16‐, cGAS‐ and STING‐dependent pathway

Kathrine Hansen; Thaneas Prabakaran; Anders Laustsen; Sofie Jørgensen; Stine H Rahbæk; Søren B. Jensen; Rikke Nielsen; Jess H. Leber; Thomas Decker; Kristy A. Horan; Martin R. Jakobsen; Søren R. Paludan

Listeria monocytogenes is a gram‐positive facultative intracellular bacterium, which replicates in the cytoplasm of myeloid cells. Interferon β (IFNβ) has been reported to play an important role in the mechanisms underlying Listeria disease. Although studies in murine cells have proposed the bacteria‐derived cyclic‐di‐AMP to be the key bacterial immunostimulatory molecule, the mechanism for IFNβ expression during L. monocytogenes infection in human myeloid cells remains unknown. Here we report that in human macrophages, Listeria DNA rather than cyclic‐di‐AMP is stimulating the IFN response via a pathway dependent on the DNA sensors IFI16 and cGAS as well as the signalling adaptor molecule STING. Thus, Listeria DNA is a major trigger of IFNβ expression in human myeloid cells and is sensed to activate a pathway dependent on IFI16, cGAS and STING.


Nature Communications | 2016

Influenza A virus targets a cGAS-independent STING pathway that controls enveloped RNA viruses

Christian K. Holm; Stine H. Rahbek; Hans Henrik Gad; Rasmus O. Bak; Martin R. Jakobsen; Zhaozaho Jiang; Anne Louise Hansen; Simon K. Jensen; Chenglong Sun; Martin K. Thomsen; Anders Laustsen; Camilla G. Nielsen; Kasper Severinsen; Yingluo Xiong; Dara L. Burdette; Veit Hornung; Robert Jan Lebbink; Mogens Duch; Katherine A. Fitzgerald; Shervin Bahrami; Jakob Giehm Mikkelsen; Rune Hartmann; Søren R. Paludan

Stimulator of interferon genes (STING) is known be involved in control of DNA viruses but has an unexplored role in control of RNA viruses. During infection with DNA viruses STING is activated downstream of cGAMP synthase (cGAS) to induce type I interferon. Here we identify a STING-dependent, cGAS-independent pathway important for full interferon production and antiviral control of enveloped RNA viruses, including influenza A virus (IAV). Further, IAV interacts with STING through its conserved hemagglutinin fusion peptide (FP). Interestingly, FP antagonizes interferon production induced by membrane fusion or IAV but not by cGAMP or DNA. Similar to the enveloped RNA viruses, membrane fusion stimulates interferon production in a STING-dependent but cGAS-independent manner. Abolishment of this pathway led to reduced interferon production and impaired control of enveloped RNA viruses. Thus, enveloped RNA viruses stimulate a cGAS-independent STING pathway, which is targeted by IAV.


Nature Communications | 2017

IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP

Kasper L. Jønsson; Anders Laustsen; Christian Krapp; K. A. Skipper; Karthiga Thavachelvam; D. Hotter; J. H. Egedal; M. Kjolby; Pejman Mohammadi; Thaneas Prabakaran; L. K. Sørensen; Chenglong Sun; Søren B. Jensen; Christian K. Holm; Robert Jan Lebbink; Mogens Johannsen; Mette Nyegaard; Jacob Giehm Mikkelsen; F. Kirchhoff; Søren R. Paludan; Martin R. Jakobsen

Innate immune activation by macrophages is an essential part of host defence against infection. Cytosolic recognition of microbial DNA in macrophages leads to induction of interferons and cytokines through activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING). Other host factors, including interferon-gamma inducible factor 16 (IFI16), have been proposed to contribute to immune activation by DNA. However, their relation to the cGAS-STING pathway is not clear. Here, we show that IFI16 functions in the cGAS-STING pathway on two distinct levels. Depletion of IFI16 in macrophages impairs cGAMP production on DNA stimulation, whereas overexpression of IFI16 amplifies the function of cGAS. Furthermore, IFI16 is vital for the downstream signalling stimulated by cGAMP, facilitating recruitment and activation of TANK-binding kinase 1 in STING complex. Collectively, our results suggest that IFI16 is essential for efficient sensing and signalling upon DNA challenge in macrophages to promote interferons and antiviral responses.


PLOS Pathogens | 2017

Mucosal stromal fibroblasts markedly enhance HIV infection of CD4+ T cells

Jason Neidleman; Joseph C. Chen; Nargis Kohgadai; Janis A. Müller; Anders Laustsen; Karthiga Thavachelvam; Karen S. Jang; Christina M. Stürzel; Jennifer J. Jones; Christina Ochsenbauer; Avantika S. Chitre; Ma Somsouk; Maurice Garcia; James F. Smith; Ruth M. Greenblatt; Jan Münch; Martin R. Jakobsen; Linda C. Giudice; Warner C. Greene; Nadia R. Roan

Understanding early events of HIV transmission within mucosal tissues is vital for developing effective prevention strategies. Here, we report that primary stromal fibroblasts isolated from endometrium, cervix, foreskin, male urethra, and intestines significantly increase HIV infection of CD4+ T cells–by up to 37-fold for R5-tropic HIV and 100-fold for X4-tropic HIV–without themselves becoming infected. Fibroblasts were more efficient than dendritic cells at trans-infection and mediate this response in the absence of the DC-SIGN and Siglec-1 receptors. In comparison, mucosal epithelial cells secrete antivirals and inhibit HIV infection. These data suggest that breaches in the epithelium allow external or luminal HIV to escape an antiviral environment to access the infection-favorable environment of the stromal fibroblasts, and suggest that resident fibroblasts have a central, but previously unrecognized, role in HIV acquisition at mucosal sites. Inhibiting fibroblast-mediated enhancement of HIV infection should be considered as a novel prevention strategy.


eLife | 2016

Targeted, homology-driven gene insertion in stem cells by ZFN-loaded ‘all-in-one’ lentiviral vectors

Yujia Cai; Anders Laustsen; Yan Zhou; Chenglong Sun; Mads Valdemar Anderson; Shengting Li; Niels Uldbjerg; Yonglun Luo; Martin R. Jakobsen; Jacob Giehm Mikkelsen

Biased integration remains a key challenge for gene therapy based on lentiviral vector technologies. Engineering of next-generation lentiviral vectors targeting safe genomic harbors for insertion is therefore of high relevance. In a previous paper (Cai et al., 2014a), we showed the use of integrase-defective lentiviral vectors (IDLVs) as carriers of complete gene repair kits consisting of zinc-finger nuclease (ZFN) proteins and repair sequences, allowing gene correction by homologous recombination (HR). Here, we follow this strategy to engineer ZFN-loaded IDLVs that insert transgenes by a homology-driven mechanism into safe loci. This insertion mechanism is driven by time-restricted exposure of treated cells to ZFNs. We show targeted gene integration in human stem cells, including CD34+ hematopoietic progenitors and induced pluripotent stem cells (iPSCs). Notably, targeted insertions are identified in 89% of transduced iPSCs. Our findings demonstrate the applicability of nuclease-loaded ‘all-in-one’ IDLVs for site-directed gene insertion in stem cell-based gene therapies. DOI: http://dx.doi.org/10.7554/eLife.12213.001


Antimicrobial Agents and Chemotherapy | 2015

Histone Deacetylase Inhibitor Romidepsin Inhibits de novo HIV-1 Infections

Kasper L. Jønsson; Martin Tolstrup; Johan Vad-Nielsen; Kathrine Kjær; Anders Laustsen; Morten Andersen; Thomas A. Rasmussen; Ole S. Søgaard; Lars Østergaard; Paul W. Denton; Martin R. Jakobsen

ABSTRACT Adjunct therapy with the histone deacetylase inhibitor (HDACi) romidepsin increases plasma viremia in HIV patients on combination antiretroviral therapy (cART). However, a potential concern is that reversing HIV latency with an HDACi may reactivate the virus in anatomical compartments with suboptimal cART concentrations, leading to de novo infection of susceptible cells in these sites. We tested physiologically relevant romidepsin concentrations known to reactivate latent HIV in order to definitively address this concern. We found that romidepsin significantly inhibited HIV infection in peripheral blood mononuclear cells and CD4+ T cells but not in monocyte-derived macrophages. In addition, romidepsin impaired HIV spreading in CD4+ T cell cultures. When we evaluated the impact of romidepsin on quantitative viral outgrowth assays with primary resting CD4+ T cells, we found that resting CD4+ T cells exposed to romidepsin exhibited reduced proliferation and viability. This significantly lowered assay sensitivity when measuring the efficacy of romidepsin as an HIV latency reversal agent. Altogether, our data indicate that romidepsin-based HIV eradication strategies are unlikely to reseed a latent T cell reservoir, even under suboptimal cART conditions, because romidepsin profoundly restricts de novo HIV infections.


Nature Communications | 2018

Nrf2 negatively regulates STING indicating a link between antiviral sensing and metabolic reprogramming

David Olagnier; Aske M. Brandtoft; Camilla Gunderstofte; Nikolaj L. Villadsen; Christian Krapp; Anne L. Thielke; Anders Laustsen; Suraj Peri; Anne Louise Hansen; Lene Bonefeld; Jacob Thyrsted; Victor Bruun; Marie B. Iversen; Lin Lin; Virginia M. Artegoitia; Chenhe Su; Long Yang; Rongtuan Lin; Siddharth Balachandran; Yonglun Luo; Mette Nyegaard; Bernadette Marrero; Raphaela Goldbach-Mansky; Mona Motwani; Dylan G. Ryan; Katherine A. Fitzgerald; Luke A.J. O’Neill; Anne Kruse Hollensen; Christian Kroun Damgaard; Frank de Paoli

The transcription factor Nrf2 is a critical regulator of inflammatory responses. If and how Nrf2 also affects cytosolic nucleic acid sensing is currently unknown. Here we identify Nrf2 as an important negative regulator of STING and suggest a link between metabolic reprogramming and antiviral cytosolic DNA sensing in human cells. Here, Nrf2 activation decreases STING expression and responsiveness to STING agonists while increasing susceptibility to infection with DNA viruses. Mechanistically, Nrf2 regulates STING expression by decreasing STING mRNA stability. Repression of STING by Nrf2 occurs in metabolically reprogrammed cells following TLR4/7 engagement, and is inducible by a cell-permeable derivative of the TCA-cycle-derived metabolite itaconate (4-octyl-itaconate, 4-OI). Additionally, engagement of this pathway by 4-OI or the Nrf2 inducer sulforaphane is sufficient to repress STING expression and type I IFN production in cells from patients with STING-dependent interferonopathies. We propose Nrf2 inducers as a future treatment option in STING-dependent inflammatory diseases.Understanding how regulators of inflammation affect nucleic acid sensing is important for targeting research against inflammatory diseases and conditions. Here, the authors identify Nrf2 as an important negative regulator of STING and suggest a link between metabolic reprogramming and antiviral cytosolic DNA sensing in human cells.


Nature Communications | 2018

Interferon priming is essential for human CD34+ cell-derived plasmacytoid dendritic cell maturation and function

Anders Laustsen; Rasmus O. Bak; Christian Krapp; L. Kjær; J. H. Egedahl; C. C. Petersen; Satish K. Pillai; H. Q. Tang; Niels Uldbjerg; Matthew H. Porteus; Nadia R. Roan; Mette Nyegaard; P. W. Denton; Martin R. Jakobsen

Plasmacytoid dendritic cells (pDC) are essential for immune competence. Here we show that pDC precursor differentiated from human CD34+ hematopoietic stem and progenitor cells (HSPC) has low surface expression of pDC markers, and has limited induction of type I interferon (IFN) and IL-6 upon TLR7 and TLR9 agonists treatment; by contrast, cGAS or RIG-I agonists-mediated activation is not altered. Importantly, after priming with type I and II IFN, these precursor pDCs attain a phenotype and functional activity similar to that of peripheral blood-derived pDCs. Data from CRISPR/Cas9-mediated genome editing of HSPCs further show that HSPC-pDCs with genetic modifications can be obtained, and that expression of the IFN-α receptor is essential for the optimal function, but dispensable for the differentiation, of HSPC-pDC percursor. Our results thus demonstrate the biological effects of IFNs for regulating pDC function, and provide the means of generating of gene-modified human pDCs.Plasmacytoid dendritic cells (pDC) are an important regulator of immune responses. Here the authors show that pDC precursors, similar to peripheral blood-derived pDCs, can be differentiated from human CD34+ hematopoietic stem and progenitor cells, with type I/II IFN priming being required for their functional maturation and differentiation.


Molecular Therapy | 2016

127. Lentiviral Protein Transduction for Tailored Genome Editing and Site-Directed Gene Insertion

Yujia Cai; Rasmus O. Bak; Anders Laustsen; Yan Zhou; Chenglong Sun; Yonglun Luo; Martin R. Jakobsen; Jacob Giehm Mikkelsen

Therapeutic use of site-directed endonucleases relies on safe and effective cellular delivery, preferentially resulting in short-term enzymatic activity. Based on the packaging of Gag/GagPol-fused heterologous proteins into VSV-G-pseudotyped lentivirus-derived particles, we have established lentiviral protein transduction for delivery of DNA transposases and custom-made endonucleases. Up to 24% of targeted CCR5 and AAVS1 alleles were disrupted in primary cells, including normal human dermal fibroblasts and primary keratinocytes, exposed to lentiviral particles loaded with zinc-finger nucleases (ZFNs). By exposing human 293 cells to ‘all-in-one’ integrase-defective lentiviral vectors (IDLVs) containing a complete gene repair kit consisting of ZFNs and viral RNA carrying the donor sequence for homology-directed repair, correction of genomic mutations was obtained in more than 8% of treated cells. As shown by confocal microscopy, ZFN proteins were abundant within transduced cells one hour after initial virus exposure, but were short-lived and gone after 24 hours. In accordance, under conditions supporting comparable CCR5 indel rates, disruption of the nearby CCR2 off-target site was reduced by lentiviral delivery of ZFNs targeting CCR5 relative to a conventional transfection-based approach. As biased and uncontrolled integration into genes remains a key challenge for gene therapies based on lentiviral vector technologies, we engineered ZFN-loaded IDLVs with the capacity to insert transgenes into the human CCR5 and AAVS1 loci by a homology-driven mechanism. Targeted gene integration into safe genomic loci was observed in human cell lines (85% of analyzed clones) and in human stem cells, including CD34+ hematopoietic progenitors and induced pluripotent stem cells (iPSCs). Notably, targeted transgene insertion into safe harbors was identified in all of 23 analyzed iPSC clones. Altogether, our findings generate a new platform for targeted genome engineering based on lentiviral delivery of complete gene repair or gene insertion kits.

Collaboration


Dive into the Anders Laustsen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katherine A. Fitzgerald

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge