Network


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

Hotspot


Dive into the research topics where Marios Koutsakos is active.

Publication


Featured researches published by Marios Koutsakos.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Human mucosal-associated invariant T cells contribute to antiviral influenza immunity via IL-18–dependent activation

Liyen Loh; Zhongfang Wang; Sneha Sant; Marios Koutsakos; Sinthujan Jegaskanda; Alexandra J. Corbett; Ligong Liu; David P. Fairlie; Jane Crowe; Jamie Rossjohn; Jianqing Xu; Peter C. Doherty; James McCluskey; Katherine Kedzierska

Significance Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes with potent antibacterial reactivity. In this study, we investigated whether MAIT cells also contribute to immunity against influenza A viruses. Compared with those who succumbed, hospitalized patients who recovered from severe avian H7N9 influenza infection had higher numbers of MAIT cells. Subsequent in vitro analysis established that MAIT cells from healthy donors are indirectly activated by influenza infection via an IL-18–dependent (but not IL-12–dependent) mechanism requiring the involvement of CD14+ monocytes. Our findings highlight the potential for MAIT cells to promote protective immunity in human influenza. Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes known to elicit potent immunity to a broad range of bacteria, mainly via the rapid production of inflammatory cytokines. Whether MAIT cells contribute to antiviral immunity is less clear. Here we asked whether MAIT cells produce cytokines/chemokines during severe human influenza virus infection. Our analysis in patients hospitalized with avian H7N9 influenza pneumonia showed that individuals who recovered had higher numbers of CD161+Vα7.2+ MAIT cells in peripheral blood compared with those who succumbed, suggesting a possible protective role for this lymphocyte population. To understand the mechanism underlying MAIT cell activation during influenza, we cocultured influenza A virus (IAV)-infected human lung epithelial cells (A549) and human peripheral blood mononuclear cells in vitro, then assayed them by intracellular cytokine staining. Comparison of influenza-induced MAIT cell activation with the profile for natural killer cells (CD56+CD3−) showed robust up-regulation of IFNγ for both cell populations and granzyme B in MAIT cells, although the individual responses varied among healthy donors. However, in contrast to the requirement for cell-associated factors to promote NK cell activation, the induction of MAIT cell cytokine production was dependent on IL-18 (but not IL-12) production by IAV-exposed CD14+ monocytes. Overall, this evidence for IAV activation via an indirect, IL-18–dependent mechanism indicates that MAIT cells are protective in influenza, and also possibly in any human disease process in which inflammation and IL-18 production occur.


Future Microbiology | 2016

Knowns and unknowns of influenza B viruses

Marios Koutsakos; Thi H. O. Nguyen; Wendy S. Barclay; Katherine Kedzierska

Influenza B viruses (IBVs) circulate annually along with influenza A (IAV) strains during seasonal epidemics. IBV can dominate influenza seasons and cause severe disease, particularly in children and adolescents. Research has revealed interesting aspects of IBV and highlighted the importance of these viruses in clinical settings. Yet, many important questions remain unanswered. In this review, the clinical relevance of IBV is emphasized, unique features in epidemiology, host range and virology are highlighted and gaps in knowledge pinpointed. Multiple aspects of IBV epidemiology, evolution, virology and immunology are discussed. Future research into IBV is needed to understand how we can prevent severe disease in high-risk groups, especially children and elderly.


Science Translational Medicine | 2018

Circulating TFH cells, serological memory, and tissue compartmentalization shape human influenza-specific B cell immunity

Marios Koutsakos; Adam K. Wheatley; Liyen Loh; E. Bridie Clemens; Sneha Sant; Simone Nüssing; Annette Fox; Amy W. Chung; Karen L. Laurie; Aeron C. Hurt; Steve Rockman; Martha Lappas; Thomas Loudovaris; Stuart I. Mannering; Glen P. Westall; Michael Elliot; Stuart G. Tangye; Linda M. Wakim; Stephen J. Kent; Thi H. O. Nguyen; Katherine Kedzierska

Analysis of influenza-specific B cells during antigen exposure and tissue compartmentalization provides insights into human B cell memory. Investigating influenza immunity Seasonal influenza vaccines have been recommended for decades, but studies focused on antigen-specific lymphocytes in humans are sparse. Koutsakos et al. examined longitudinal samples of influenza-vaccinated individuals to determine what responses generate protective immunity. Vaccination could induce circulating T follicular helper memory cells, antibody-secreting cells, and memory B cells, but did not seem to affect other types of lymphocytes. Existing anti-influenza antibodies at the time of vaccination dampened these responses. They probed different types of human tissues to hunt for influenza memory B cells, thereby showing that the memory response exists outside the circulation. Better targeting these cells could improve influenza vaccine efficacy. Immunization with the inactivated influenza vaccine (IIV) remains the most effective strategy to combat seasonal influenza infections. IIV activates B cells and T follicular helper (TFH) cells and thus engenders antibody-secreting cells and serum antibody titers. However, the cellular events preceding generation of protective immunity in humans are inadequately understood. We undertook an in-depth analysis of B cell and T cell immune responses to IIV in 35 healthy adults. Using recombinant hemagglutinin (rHA) probes to dissect the quantity, phenotype, and isotype of influenza-specific B cells against A/California09-H1N1, A/Switzerland-H3N2, and B/Phuket, we showed that vaccination induced a three-pronged B cell response comprising a transient CXCR5−CXCR3+ antibody-secreting B cell population, CD21hiCD27+ memory B cells, and CD21loCD27+ B cells. Activation of circulating TFH cells correlated with the development of both CD21lo and CD21hi memory B cells. However, preexisting antibodies could limit increases in serum antibody titers. IIV had no marked effect on CD8+, mucosal-associated invariant T, γδ T, and natural killer cell activation. In addition, vaccine-induced B cells were not maintained in peripheral blood at 1 year after vaccination. We provide a dissection of rHA-specific B cells across seven human tissue compartments, showing that influenza-specific memory (CD21hiCD27+) B cells primarily reside within secondary lymphoid tissues and the lungs. Our study suggests that a rational design of universal vaccines needs to consider circulating TFH cells, preexisting serological memory, and tissue compartmentalization for effective B cell immunity, as well as to improve targeting cellular T cell immunity.


Frontiers of Medicine in China | 2018

Innate and adaptive T cells in influenza disease

Simone Nüssing; Sneha Sant; Marios Koutsakos; Kanta Subbarao; Thi H. O. Nguyen; Katherine Kedzierska

Influenza is a major global health problem, causing infections of the respiratory tract, often leading to acute pneumonia, life-threatening complications and even deaths. Over the last seven decades, vaccination strategies have been utilized to protect people from complications of influenza, especially groups at high risk of severe disease. While current vaccination regimens elicit strain-specific antibody responses, they fail to generate cross-protection against seasonal, pandemic and avian viruses. Moreover, vaccines designed to generate influenza-specific T-cell responses are yet to be optimized. During natural infection, viral replication is initially controlled by innate immunity before adaptive immune responses (T cells and antibody-producing B cells) achieve viral clearance and host recovery. Adaptive T and B cells maintain immunological memory and provide protection against subsequent infections with related influenza viruses. Recent studies also shed light on the role of innate T-cells (MAIT cells, γδ cells, and NKT cells) in controlling influenza and linking innate and adaptive immune mechanisms, thus making them attractive targets for vaccination strategies. We summarize the current knowledge on influenza-specific innate MAIT and γδ T cells as well as adaptive CD8+ and CD4+ T cells, and discuss how these responses can be harnessed by novel vaccine strategies to elicit cross-protective immunity against different influenza strains and subtypes.


Journal of Leukocyte Biology | 2017

Perturbed CD8+ T cell immunity across universal influenza epitopes in the elderly

Thi H. O. Nguyen; Sneha Sant; Nicola L. Bird; Emma J. Grant; E. Bridie Clemens; Marios Koutsakos; Sophie A. Valkenburg; Stephanie Gras; Martha Lappas; Anthony Jaworowski; Jane Crowe; Liyen Loh; Katherine Kedzierska

Influenza epidemics lead to severe illness, life‐threatening complications, and deaths, especially in the elderly. As CD8+ T cells are associated with rapid recovery from influenza, we investigated the effects of aging on antigen‐specific CD8+ T cells across the universal influenza epitopes in humans. We show that aging is characterized by altered frequencies in T cell subsets, with naive T cells being partially replaced by activated effector/memory populations. Although we observed no striking differences in TCR signaling capacity, T cells in the elderly had increased expression of transcription factors Eomes and T‐bet, and such changes were most apparent in CD8+ T cells. Strikingly, the numbers of antigen‐specific CD8+ T cells across universal influenza epitopes were reduced in the elderly, although their effector/memory phenotypes remained stable. To understand whether diminished numbers of influenza‐specific CD8+ T cells in the elderly resulted from alteration in TCR clonotypes, we dissected the TCRαβ repertoire specific for the prominent HLA‐A*02:01‐restricted‐M158–66 (A2/M158) influenza epitope. We provide the first ex vivo data on paired antigen‐specific TCRαβ clonotypes in the elderly, showing that influenza‐specific A2/M158+ TCRαβ repertoires in the elderly adults varied from those in younger adults, with the main features being a reduction in the frequency of the public TRAV27–TRBV19 TCRαβ clonotype, increased proportion of private TCRαβ signatures, broader use of TRAV and TRBV gene segments, and large clonal expansion of private TCRαβ clonotypes with longer CDR3 loops. Our study supports the development of T cell‐targeted influenza vaccines that would boost the T cell compartment during life and maintain the numbers and optimal TCRαβ signatures in the elderly.


Nature Communications | 2018

Clonally diverse CD38 + HLA-DR + CD8 + T cells persist during fatal H7N9 disease

Zhongfang Wang; Lingyan Zhu; Thi H. O. Nguyen; Yanmin Wan; Sneha Sant; Sergio Quiñones-Parra; Jeremy Chase Crawford; Auda A. Eltahla; Simone Rizzetto; Rowena A. Bull; Chenli Qiu; Marios Koutsakos; E. Bridie Clemens; Liyen Loh; Tianyue Chen; Lu Liu; Pengxing Cao; Yanqin Ren; Lukasz Kedzierski; Tom Kotsimbos; James M. McCaw; Nicole L. La Gruta; Stephen J. Turner; Allen C. Cheng; Fabio Luciani; Xiaoyan Zhang; Peter C. Doherty; Paul G. Thomas; Jianqing Xu; Katherine Kedzierska

Severe influenza A virus (IAV) infection is associated with immune dysfunction. Here, we show circulating CD8+ T-cell profiles from patients hospitalized with avian H7N9, seasonal IAV, and influenza vaccinees. Patient survival reflects an early, transient prevalence of highly activated CD38+HLA-DR+PD-1+ CD8+ T cells, whereas the prolonged persistence of this set is found in ultimately fatal cases. Single-cell T cell receptor (TCR)-αβ analyses of activated CD38+HLA-DR+CD8+ T cells show similar TCRαβ diversity but differential clonal expansion kinetics in surviving and fatal H7N9 patients. Delayed clonal expansion associated with an early dichotomy at a transcriptome level (as detected by single-cell RNAseq) is found in CD38+HLA-DR+CD8+ T cells from patients who succumbed to the disease, suggesting a divergent differentiation pathway of CD38+HLA-DR+CD8+ T cells from the outset during fatal disease. Our study proposes that effective expansion of cross-reactive influenza-specific TCRαβ clonotypes with appropriate transcriptome signatures is needed for early protection against severe influenza disease.Virus-specific CD8+ T cells are crucial during H7N9 influenza infection, but CD8+ T cell dysfunction is associated with poor prognosis. Here, the authors use molecular and phenotypic analysis to establish persistence of clonally diverse CD8+ T cell populations during fatal infection.


ACS Chemical Biology | 2017

Augmenting Influenza-Specific T Cell Memory Generation with a Natural Killer T Cell-Dependent Glycolipid–Peptide Vaccine

Regan J. Anderson; Jasmine Li; Lukasz Kedzierski; Benjamin J. Compton; Colin M. Hayman; Taryn L. Osmond; Ching Wen Tang; Kathryn J. Farrand; Hui Fern Koay; Catarina F Almeida; Lauren R. Holz; Geoffrey M. Williams; Margaret A. Brimble; Zhongfang Wang; Marios Koutsakos; Katherine Kedzierska; Dale I. Godfrey; Ian F. Hermans; Stephen J. Turner; Gavin F. Painter

The development of a universal vaccine for influenza A virus (IAV) that does not require seasonal modification is a long-standing health goal, particularly in the context of the increasing threat of new global pandemics. Vaccines that specifically induce T cell responses are of considerable interest because they can target viral proteins that are more likely to be shared between different virus strains and subtypes and hence provide effective cross-reactive IAV immunity. From a practical perspective, such vaccines should induce T cell responses with long-lasting memory, while also being simple to manufacture and cost-effective. Here we describe the synthesis and evaluation of a vaccine platform based on solid phase peptide synthesis and bio-orthogonal conjugation methodologies. The chemical approach involves covalently attaching synthetic long peptides from a virus-associated protein to a powerful adjuvant molecule, α-galactosylceramide (α-GalCer). Strain-promoted azide-alkyne cycloaddition is used as a simple and efficient method for conjugation, and pseudoproline methodology is used to increase the efficiency of the peptide synthesis. α-GalCer is a glycolipid that stimulates NKT cells, a population of lymphoid-resident immune cells that can provide potent stimulatory signals to antigen-presenting cells engaged in driving proliferation and differentiation of peptide-specific T cells. When used in mice, the vaccine induced T cell responses that provided effective prophylactic protection against IAV infection, with the speed of viral clearance greater than that seen from previous viral exposure. These findings are significant because the vaccines are highly defined, quick to synthesize, and easily characterized and are therefore appropriate for large scale affordable manufacture.


bioRxiv | 2018

MAIT cells contribute to protection against lethal influenza infection in vivo

Bonnie van Wilgenburg; Liyen Loh; Zhenjun Chen; Troi Pediongco; Huimeng Wang; Mai Shi; Zhe Zhao; Marios Koutsakos; Simone Nüssing; Sneha Sant; Zhongfang Wang; Criselle D'Souza; Catarina F Almeida; Lyudmila Kostenko; Sidonia B. G. Eckle; Bronwyn Meehan; Dale I. Godfrey; Patrick C. Reading; Alexandra J. Corbett; James McCluskey; Paul Klenerman; Katherine Kedzierska; Timothy S. C. Hinks

Mucosal associated invariant T (MAIT) cells are evolutionarily-conserved, innate-like lymphocytes which are abundant in human lungs and can contribute to protection against pulmonary bacterial infection. MAIT cells are also activated during human viral infections, yet it remains unknown whether MAIT cells play a significant protective or even detrimental role during viral infections in vivo. Using murine experimental challenge with two strains of influenza A virus, we show that MAIT cells accumulated and were activated early in infection, with upregulation of CD25, CD69 and Granzyme B, peaking at 5 days post infection. Activation was modulated via cytokines independently of MR1. MAIT cell-deficient MR1−/− mice showed enhanced weight loss and mortality to severe (H1N1) influenza. This was ameliorated by prior adoptive transfer of pulmonary MAIT cells in both immunocompetent and immunodeficient RAG2−/−γC−/− mice. Thus, MAIT cells contribute to protection during respiratory viral infections, and constitute a potential target for therapeutic manipulation.


Journal of General Virology | 2016

NB protein does not affect influenza B virus replication in vitro and is not required for replication in or transmission between ferrets

Ruth A. Elderfield; Marios Koutsakos; Rebecca Frise; Konrad Bradley; Jonathan W. Ashcroft; Shanhjahan Miah; Angie Lackenby; Wendy S. Barclay

The influenza B virus encodes a unique protein, NB, a membrane protein whose function in the replication cycle is not, as yet, understood. We engineered a recombinant influenza B virus lacking NB expression, with no concomitant difference in expression or activity of viral neuraminidase (NA) protein, an important caveat since NA is encoded on the same segment and initiated from a start codon just 4 nt downstream of NB. Replication of the virus lacking NB was not different to wild-type virus with full-length NB in clonal immortalized or complex primary cell cultures. In the mouse model, virus lacking NB induced slightly lower IFN-α levels in infected lungs, but this did not affect virus titres or weight loss. In ferrets infected with a mixture of viruses that did or did not express NB, there was no fitness advantage for the virus that retained NB. Moreover, virus lacking NB protein was transmitted following respiratory droplet exposure of sentinel animals. These data suggest no role for NB in supporting replication or transmission in vivo in this animal model. The role of NB and the nature of selection to retain it in all natural influenza B viruses remain unclear.


Frontiers in Immunology | 2018

Single-Cell Approach to Influenza-Specific CD8+ T Cell Receptor Repertoires Across Different Age Groups, Tissues, and Following Influenza Virus Infection

Sneha Sant; Ludivine Grzelak; Zhongfang Wang; Angela Pizzolla; Marios Koutsakos; Jane Crowe; Thomas Loudovaris; Stuart I. Mannering; Glen P. Westall; Linda M. Wakim; Jamie Rossjohn; Stephanie Gras; Michael J. Richards; Jianqing Xu; Paul G. Thomas; Liyen Loh; Thi H. O. Nguyen; Katherine Kedzierska

CD8+ T cells recognizing antigenic peptides derived from conserved internal viral proteins confer broad protection against distinct influenza viruses. As memory CD8+ T cells change throughout the human lifetime and across tissue compartments, we investigated how T cell receptor (TCR) composition and diversity relate to memory CD8+ T cells across anatomical sites and immunological phases of human life. We used ex vivo peptide-HLA tetramer magnetic enrichment, single-cell multiplex RT-PCR for both the TCR-alpha (TCRα) and TCR-beta (TCRβ) chains, and new TCRdist and grouping of lymphocyte interactions by paratope hotspots (GLIPH) algorithms to compare TCRs directed against the most prominent human influenza epitope, HLA-A*02:01-M158–66 (A2+M158). We dissected memory TCR repertoires directed toward A2+M158 CD8+ T cells within human tissues and compared them to human peripheral blood of young and elderly adults. Furthermore, we compared these memory CD8+ T cell repertoires to A2+M158 CD8+ TCRs during acute influenza disease in patients hospitalized with avian A/H7N9 virus. Our study provides the first ex vivo comparative analysis of paired antigen-specific TCR-α/β clonotypes across different tissues and peripheral blood across different age groups. We show that human A2+M158 CD8+ T cells can be readily detected in human lungs, spleens, and lymph nodes, and that tissue A2+M158 TCRαβ repertoires reflect A2+M158 TCRαβ clonotypes derived from peripheral blood in healthy adults and influenza-infected patients. A2+M158 TCRαβ repertoires displayed distinct features only in elderly adults, with large private TCRαβ clonotypes replacing the prominent and public TRBV19/TRAV27 TCRs. Our study provides novel findings on influenza-specific TCRαβ repertoires within human tissues, raises the question of how we can prevent the loss of optimal TCRαβ signatures with aging, and provides important insights into the rational design of T cell-mediated vaccines and immunotherapies.

Collaboration


Dive into the Marios Koutsakos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sneha Sant

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liyen Loh

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge