Network


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

Hotspot


Dive into the research topics where Jakob Michaëlsson is active.

Publication


Featured researches published by Jakob Michaëlsson.


Science | 2008

Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero.

Jeff E. Mold; Jakob Michaëlsson; Trevor D. Burt; Marcus O. Muench; Karen P. Beckerman; Michael P. Busch; Tzong-Hae Lee; Douglas F. Nixon; Joseph M. McCune

As the immune system develops, T cells are selected or regulated to become tolerant of self antigens and reactive against foreign antigens. In mice, the induction of such tolerance is thought to be attributable to the deletion of self-reactive cells. Here, we show that the human fetal immune system takes advantage of an additional mechanism: the generation of regulatory T cells (Tregs) that suppress fetal immune responses. We find that substantial numbers of maternal cells cross the placenta to reside in fetal lymph nodes, inducing the development of CD4+CD25highFoxP3+ Tregs that suppress fetal antimaternal immunity and persist at least until early adulthood. These findings reveal a form of antigen-specific tolerance in humans, induced in utero and probably active in regulating immune responses after birth.


Journal of Virology | 2004

Human CD4+ CD25+ Regulatory T Cells Control T-Cell Responses to Human Immunodeficiency Virus and Cytomegalovirus Antigens

Einar Martin Aandahl; Jakob Michaëlsson; Walter J. Moretto; Frederick Hecht; Douglas F. Nixon

ABSTRACT Regulatory T (TR) cells maintain tolerance to self-antigens and control immune responses to alloantigens after organ transplantation. Here, we show that CD4+ CD25+ human TR cells suppress virus-specific T-cell responses. Depletion of TR cells from peripheral blood mononuclear cells enhances T-cell responses to cytomegalovirus and human immunodeficiency virus antigens. We propose that chronic viral infections lead to induction of suppressive TR cells that inhibit the antiviral immune response.


Blood | 2010

Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK cell differentiation uncoupled from NK cell education

Niklas K. Björkström; Peggy Riese; Frank Heuts; Sandra Andersson; Cyril Fauriat; Martin A. Ivarsson; Andreas Björklund; Malin Flodström-Tullberg; Jakob Michaëlsson; Martin E. Rottenberg; Carlos A. Guzmán; Hans-Gustaf Ljunggren; Karl-Johan Malmberg

Natural killer (NK) cells are lymphocytes of the innate immune system that, following differentiation from CD56(bright) to CD56(dim) cells, have been thought to retain fixed functional and phenotypic properties throughout their lifespan. In contrast to this notion, we here show that CD56(dim) NK cells continue to differentiate. During this process, they lose expression of NKG2A, sequentially acquire inhibitory killer cell inhibitory immunoglobulin-like receptors and CD57, change their expression patterns of homing molecules, and display a gradual decline in proliferative capacity. All cellular intermediates of this process are represented in varying proportions at steady state and appear, over time, during the reconstitution of the immune system, as demonstrated in humanized mice and in patients undergoing hematopoietic stem cell transplantation. CD56(dim) NK-cell differentiation, and the associated functional imprint, occurs independently of NK-cell education by interactions with self-human leukocyte antigen class I ligands and is an essential part of the formation of human NK-cell repertoires.


Journal of Experimental Medicine | 2011

Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus

Niklas K. Björkström; Therese Lindgren; Malin Stoltz; Cyril Fauriat; Monika Braun; Magnus Evander; Jakob Michaëlsson; Karl-Johan Malmberg; Jonas Klingström; Clas Ahlm; Hans-Gustaf Ljunggren

Acute hantavirus infection in humans triggers a rapid expansion and long-term persistence of NK cells.


Science | 2010

Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans.

Jeff E. Mold; Shivkumar Venkatasubrahmanyam; Trevor D. Burt; Jakob Michaëlsson; Jose M. Rivera; Sofiya A. Galkina; Kenneth I. Weinberg; Cheryl A. Stoddart; Joseph M. McCune

Lymphocytes Layer It On Cells of the immune system begin to develop from hematopoietic stem cells (HSCs) during fetal life. In the adult, HSCs continue to produce immune cells to replenish dying cells or in response to an infection. In mice and birds, immune cell development occurs in a “layered” manner, whereby distinct populations of HSCs that arise at different times during development generate distinct immune cell lineages. In contrast, development of human immune cells, and T lymphocytes in particular, is thought to be linear. Mold et al. (p. 1695; see the Perspective by Betz) now show that T lymphocyte development in humans is also “layered,” and strategically so. T cells that arise from fetal HSCs are enriched in regulatory T cells, which promote immune tolerance, rather than classical T cells, which readily respond to foreign antigen. By favoring the development of regulatory T cell populations during fetal life, the immune system is perhaps better able to keep responses to maternal antigens in check. The development of large numbers of classical T cells is delayed until after birth when infectious agents represent a more imminent threat. Distinct fetal T cell lineages help explain the tolerogenic properties of the fetus and immune responsiveness at birth. Although the mammalian immune system is generally thought to develop in a linear fashion, findings in avian and murine species argue instead for the developmentally ordered appearance (or “layering”) of distinct hematopoietic stem cells (HSCs) that give rise to distinct lymphocyte lineages at different stages of development. Here we provide evidence of an analogous layered immune system in humans. Our results suggest that fetal and adult T cells are distinct populations that arise from different populations of HSCs that are present at different stages of development. We also provide evidence that the fetal T cell lineage is biased toward immune tolerance. These observations offer a mechanistic explanation for the tolerogenic properties of the developing fetus and for variable degrees of immune responsiveness at birth.


Journal of Experimental Medicine | 2002

A Signal Peptide Derived from hsp60 Binds HLA-E and Interferes with CD94/NKG2A Recognition

Jakob Michaëlsson; Cristina Teixeira de Matos; Adnane Achour; Lewis L. Lanier; Klas Kärre; Kalle Söderström

Human histocompatibility leukocyte antigen (HLA)-E is a nonclassical major histocompatibility complex (MHC) class I molecule which presents a restricted set of nonameric peptides, derived mainly from the signal sequence of other MHC class I molecules. It interacts with CD94/NKG2 receptors expressed on the surface of natural killer (NK) cells and T cell subsets. Here we demonstrate that HLA-E also presents a peptide derived from the leader sequence of human heat shock protein 60 (hsp60). This peptide gains access to HLA-E intracellularly, resulting in up-regulated HLA-E/hsp60 signal peptide cell-surface levels on stressed cells. Notably, HLA-E molecules in complex with the hsp60 signal peptide are no longer recognized by CD94/NKG2A inhibitory receptors. Thus, during cellular stress an increased proportion of HLA-E molecules may bind the nonprotective hsp60 signal peptide, leading to a reduced capacity to inhibit a major NK cell population. Such stress induced peptide interference would gradually uncouple CD94/NKG2A inhibitory recognition and provide a mechanism for NK cells to detect stressed cells in a peptide-dependent manner.


Blood | 2010

Education of human natural killer cells by activating killer cell immunoglobulin-like receptors

Cyril Fauriat; Martin A. Ivarsson; Hans-Gustaf Ljunggren; Karl-Johan Malmberg; Jakob Michaëlsson

Expression of inhibitory killer cell immunoglobulin-like receptors (KIRs) specific for self-major histocompatibility complex (MHC) class I molecules provides an educational signal that generates functional natural killer (NK) cells. However, the effects of activating KIRs specific for self-MHC class I on NK-cell education remain elusive. Here, we provide evidence that the activating receptor KIR2DS1 tunes down the responsiveness of freshly isolated human NK cells to target cell stimulation in donors homozygous for human leukocyte antigen (HLA)-C2, the ligand of KIR2DS1. The tuning was apparent in KIR2DS1(+) NK cells lacking expression of inhibitory KIRs and CD94/NKG2A, as well as in KIR2DS1(+) NK cells coexpressing the inhibitory MHC class I-specific receptors CD94/NKG2A and KIR2DL3, but not KIR2DL1. However, the tuning of responsiveness was restricted to target cell recognition because KIR2DS1(+) NK cells responded well to stimulation with exogenous cytokines. Our results provide the first example of human NK-cell education by an activating KIR and suggest that the education of NK cells via activating KIRs is a mechanism to secure tolerance that complements education via inhibitory KIRs.


Journal of Immunology | 2006

Regulation of T Cell Responses in the Developing Human Fetus

Jakob Michaëlsson; Jeff E. Mold; Joseph M. McCune; Douglas F. Nixon

Although human T cells enter the peripheral lymphoid tissues early during fetal development, the adaptive immune system in the fetus has largely been regarded as functionally immature and unresponsive to stimulation. In this study, we show that depletion of fetal CD4+CD25high T regulatory (TReg) cells, which are present at high frequency in fetal lymphoid tissues, results in vigorous T cell proliferation and cytokine production in vitro, even in the absence of exogenous stimulation. Analysis of CD4+ and CD8+ T cell populations revealed a large subset of cells that expressed the early activation Ag, CD69. We show that this population represents a subset of highly reactive fetal T cells actively suppressed by fetal CD4+CD25high TReg cells during development. These findings indicate that fetal T cells are, in the absence of CD4+CD25high TReg cells, highly responsive to stimulation and provide evidence for an important role for CD4+CD25high TReg cells in controlling T cell responses in utero.


Journal of Immunology | 2007

Cutting Edge: KIR3DS1, a Gene Implicated in Resistance to Progression to AIDS, Encodes a DAP12-Associated Receptor Expressed on NK Cells That Triggers NK Cell Activation

William H. Carr; David B. Rosen; Hisashi Arase; Douglas F. Nixon; Jakob Michaëlsson; Lewis L. Lanier

The killer cell Ig-like receptor (KIR) gene, KIR3DS1, has been implicated in slowing disease progression in HIV infection; however, little is known about its expression, function, or ligand specificity. Using retrovirally transduced NKL cells and peripheral blood NK cells from KIR3DS1-positive donors we assessed expression of this gene by flow cytometry and its function by in vitro assays measuring KIR3DS1-induced cell-mediated cytotoxicity and cytokine production. In the present study, we demonstrate that KIR3DS1 is expressed on peripheral blood NK cells and triggers both cytotoxicity and IFN-γ production. Using cotransfection and coimmunoprecipitation, we found that KIR3DS1 associates with the ITAM-bearing adaptor, DAP12. Soluble KIR3DS1-Ig fusion proteins did not bind to EBV-transformed B lymphoid cell lines transfected with HLA-Bw4 80I or 80T allotypes, suggesting that if KIR3DS1 does recognize HLA-Bw4 ligands, this may be peptide dependent.


Journal of Immunology | 2000

Emergence of CD8 + T Cells Expressing NK Cell Receptors in Influenza A Virus-Infected Mice

Taku Kambayashi; Erika Assarsson; Jakob Michaëlsson; Peter Berglund; Alexander D. Diehl; Benedict J. Chambers; Hans-Gustaf Ljunggren

Both innate and adaptive immune responses play an important role in the recovery of the host from viral infections. In the present report, a subset of cells coexpressing CD8 and NKR-P1C (NK1.1) was found in the lungs of mice infected with influenza A virus. These cells were detected at low numbers in the lungs of uninfected mice, but represented up to 10% of the total CD8+ T cell population at day 10 postinfection. Almost all of the CD8+NK1.1+ cells were CD8αβ+CD3+TCRαβ+ and a proportion of these cells also expressed the NK cell-associated Ly49 receptors. Interestingly, up to 30% of these cells were virus-specific T cells as determined by MHC class I tetramer staining and by intracellular staining of IFN-γ after viral peptide stimulation. Moreover, these cells were distinct from conventional NKT cells as they were also found at increased numbers in influenza-infected CD1−/− mice. These results demonstrate that a significant proportion of CD8+ T cells acquire NK1.1 and other NK cell-associated molecules, and suggests that these receptors may possibly regulate CD8+ T cell effector functions during viral infection.

Collaboration


Dive into the Jakob Michaëlsson's collaboration.

Top Co-Authors

Avatar

Hans-Gustaf Ljunggren

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Douglas F. Nixon

George Washington University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin A. Ivarsson

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Johan K. Sandberg

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Niklas K. Björkström

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian R. Long

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge