Network


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

Hotspot


Dive into the research topics where Mikael Sigvardsson is active.

Publication


Featured researches published by Mikael Sigvardsson.


Cell | 2005

Identification of Flt3 + Lympho-Myeloid Stem Cells Lacking Erythro-Megakaryocytic Potential: A Revised Road Map for Adult Blood Lineage Commitment

Jörgen Adolfsson; Robert Månsson; Natalija Buza-Vidas; Anne Hultquist; Karina Liuba; Christina T. Jensen; David Bryder; Liping Yang; Ole-Johan Borge; Lina Thorén; Kristina Anderson; Ewa Sitnicka; Yutaka Sasaki; Mikael Sigvardsson; Sten Eirik W. Jacobsen

All blood cell lineages derive from a common hematopoietic stem cell (HSC). The current model implicates that the first lineage commitment step of adult pluripotent HSCs results in a strict separation into common lymphoid and common myeloid precursors. We present evidence for a population of cells which, although sustaining a high proliferative and combined lympho-myeloid differentiation potential, have lost the ability to adopt erythroid and megakaryocyte lineage fates. Cells in the Lin-Sca-1+c-kit+ HSC compartment coexpressing high levels of the tyrosine kinase receptor Flt3 sustain granulocyte, monocyte, and B and T cell potentials but in contrast to Lin-Sca-1+c-kit+Flt3- HSCs fail to produce significant erythroid and megakaryocytic progeny. This distinct lineage restriction site is accompanied by downregulation of genes for regulators of erythroid and megakaryocyte development. In agreement with representing a lymphoid primed progenitor, Lin-Sca-1+c-kit+CD34+Flt3+ cells display upregulated IL-7 receptor gene expression. Based on these observations, we propose a revised road map for adult blood lineage development.


Cell Stem Cell | 2007

Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy

Cornelis J.H. Pronk; Derrick J. Rossi; Robert Månsson; Joanne L. Attema; Gudmundur L. Norddahl; Charles K. Chan; Mikael Sigvardsson; Irving L. Weissman; David Bryder

The major myeloid blood cell lineages are generated from hematopoietic stem cells by differentiation through a series of increasingly committed progenitor cells. Precise characterization of intermediate progenitors is important for understanding fundamental differentiation processes and a variety of disease states, including leukemia. Here, we evaluated the functional in vitro and in vivo potentials of a range of prospectively isolated myeloid precursors with differential expression of CD150, Endoglin, and CD41. Our studies revealed a hierarchy of myeloerythroid progenitors with distinct lineage potentials. The global gene expression signatures of these subsets were consistent with their functional capacities, and hierarchical clustering analysis suggested likely lineage relationships. These studies provide valuable tools for understanding myeloid lineage commitment, including isolation of an early erythroid-restricted precursor, and add to existing models of hematopoietic differentiation by suggesting that progenitors of the innate and adaptive immune system can separate late, following the divergence of megakaryocytic/erythroid potential.


Nature Immunology | 2010

A global network of transcription factors, involving E2A, EBF1 and Foxo1, that orchestrates B cell fate

Yin C. Lin; Suchit Jhunjhunwala; Christopher Benner; Sven Heinz; Eva Welinder; Robert Månsson; Mikael Sigvardsson; James Hagman; Celso A. Espinoza; Janusz Dutkowski; Trey Ideker; Christopher K. Glass; Cornelis Murre

It is now established that the transcription factors E2A, EBF1 and Foxo1 have critical roles in B cell development. Here we show that E2A and EBF1 bound regulatory elements present in the Foxo1 locus. E2A and EBF1, as well as E2A and Foxo1, in turn, were wired together by a vast spectrum of cis-regulatory sequences. These associations were dynamic during developmental progression. Occupancy by the E2A isoform E47 directly resulted in greater abundance, as well as a pattern of monomethylation of histone H3 at lysine 4 (H3K4) across putative enhancer regions. Finally, we divided the pro-B cell epigenome into clusters of loci with occupancy by E2A, EBF and Foxo1. From this analysis we constructed a global network consisting of transcriptional regulators, signaling and survival factors that we propose orchestrates B cell fate.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion

Isabel Beerman; Deepta Bhattacharya; Sasan Zandi; Mikael Sigvardsson; Irving L. Weissman; David Bryder; Derrick J. Rossi

Aging of the hematopoietic stem cell compartment is believed to contribute to the onset of a variety of age-dependent blood cell pathophysiologies. Mechanistic drivers of hematopoietic stem cell (HSC) aging include DNA damage accumulation and induction of tumor suppressor pathways that combine to reduce the regenerative capacity of aged HSCs. Such mechanisms do not however account for the change in lymphoid and myeloid lineage potential characteristic of HSC aging, which is believed to be central to the decline of immune competence and predisposition to myelogenous diseases in the elderly. Here we have prospectively isolated functionally distinct HSC clonal subtypes, based on cell surface phenotype, bearing intrinsically different capacities to differentiate toward lymphoid and myeloid effector cells mediated by quantitative differences in lineage priming. Finally, we present data supporting a model in which clonal expansion of a class of intrinsically myeloid-biased HSCs with robust self-renewal potential is a central component of hematopoietic aging.


Immunity | 2008

E2A proteins promote development of lymphoid-primed multipotent progenitors

Sheila Dias; Robert Månsson; Sandeep Gurbuxani; Mikael Sigvardsson; Barbara L. Kee

The first lymphoid-restricted progeny of hematopoietic stem cells (HSCs) are lymphoid-primed multipotent progenitors (LMPPs), which have little erythromyeloid potential but retain lymphoid, granulocyte, and macrophage differentiation capacity. Despite recent advances in the identification of LMPPs, the transcription factors essential for their generation remain to be identified. Here, we demonstrated that the E2A transcription factors were required for proper development of LMPPs. Within HSCs and LMPPs, E2A proteins primed expression of a subset of lymphoid-associated genes and prevented expression of genes that are not normally prevalent in these cells, including HSC-associated and nonlymphoid genes. E2A proteins also restricted proliferation of HSCs, MPPs, and LMPPs and antagonized differentiation of LMPPs toward the myeloid fate. Our results reveal that E2A proteins play a critical role in supporting lymphoid specification from HSCs and that the reduced generation of LMPPs underlies the severe lymphocyte deficiencies observed in E2A-deficient mice.


Journal of Experimental Medicine | 2003

Complementary signaling through flt3 and interleukin-7 receptor alpha is indispensable for fetal and adult B cell genesis

Ewa Sitnicka; Cord Brakebusch; Inga-Lill Mårtensson; Marcus Svensson; William W. Agace; Mikael Sigvardsson; Natalija Buza-Vidas; David Bryder; Corrado M. Cilio; Henrik Ahlenius; Eugene Maraskovsky; Jacques J. Peschon; Sten Eirik W. Jacobsen

Extensive studies of mice deficient in one or several cytokine receptors have failed to support an indispensable role of cytokines in development of multiple blood cell lineages. Whereas B1 B cells and Igs are sustained at normal levels throughout life of mice deficient in IL-7, IL-7Rα, common cytokine receptor gamma chain, or flt3 ligand (FL), we report here that adult mice double deficient in IL-7Rα and FL completely lack visible LNs, conventional IgM+ B cells, IgA+ plasma cells, and B1 cells, and consequently produce no Igs. All stages of committed B cell progenitors are undetectable in FL−/− × IL-7Rα−/− BM that also lacks expression of the B cell commitment factor Pax5 and its direct target genes. Furthermore, in contrast to IL-7Rα−/− mice, FL−/− × IL-7Rα−/− mice also lack mature B cells and detectable committed B cell progenitors during fetal development. Thus, signaling through the cytokine tyrosine kinase receptor flt3 and IL-7Rα are indispensable for fetal and adult B cell development.


Nature Immunology | 2006

Intrinsic inhibition of transcription factor E2A by HLH proteins ABF-1 and Id2 mediates reprogramming of neoplastic B cells in Hodgkin lymphoma

Stephan Mathas; Martin Janz; Franziska Hummel; Michael Hummel; Brigitte Wollert-Wulf; Simone Lusatis; Ioannis Anagnostopoulos; Andreas Lietz; Mikael Sigvardsson; Franziska Jundt; Korinna Jöhrens; Kurt Bommert; Harald Stein; Bernd Dörken

B cell differentiation is controlled by a complex network of lineage-restricted transcription factors. How perturbations to this network alter B cell fate remains poorly understood. Here we show that classical Hodgkin lymphoma tumor cells, which originate from mature B cells, have lost the B cell phenotype as a result of aberrant expression of transcriptional regulators. The B cell–specific transcription factor program was disrupted by overexpression of the helix-loop-helix proteins ABF-1 and Id2. Both factors antagonized the function of the B cell–determining transcription factor E2A. As a result, expression of genes specific to B cells was lost and expression of genes not normally associated with the B lineage was upregulated. These data demonstrate the plasticity of mature human lymphoid cells and offer an explanation for the unique classical Hodgkin lymphoma phenotype.* NOTE: In the version of this article initially published online, the directions to the panels for Figure 6e were incorrect in the legend and text. The legend for this panel should begin as follows: “Immunoblot (top), EMSA (bottom left) and RT-PCR (bottom right)….” The accompanying text should read as follows: “Transfection of L428 cells with a combination of these siRNAs efficiently reduced ABF-1 protein expression (Fig. 6e, top) and resulted in a substantial loss of E2A–ABF-1 DNA-binding activity (Fig. 6e, bottom left). After reduction of ABF-1 expression, we noted considerable downregulation of CSF1R and TCF7 expression and a moderate suppression of GATA3 expression (Fig. 6e, bottom right).” The error has been corrected for the HTML and print versions of the article.


Cell Stem Cell | 2011

Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging.

Gudmundur L. Norddahl; Cornelis J.H. Pronk; Martin Wahlestedt; Gerd Sten; Jens Martin Nygren; Amol Ugale; Mikael Sigvardsson; David Bryder

Somatic stem cells mediate tissue maintenance for the lifetime of an organism. Despite the well-established longevity that is a prerequisite for such function, accumulating data argue for compromised stem cell function with age. Identifying the mechanisms underlying age-dependent stem cell dysfunction is therefore key to understanding the aging process. Here, using a model carrying a proofreading-defective mitochondrial DNA polymerase, we demonstrate hematopoietic defects reminiscent of premature HSC aging, including anemia, lymphopenia, and myeloid lineage skewing. However, in contrast to physiological stem cell aging, rapidly accumulating mitochondrial DNA mutations had little functional effect on the hematopoietic stem cell pool, and instead caused distinct differentiation blocks and/or disappearance of downstream progenitors. These results show that intact mitochondrial function is required for appropriate multilineage stem cell differentiation, but argue against mitochondrial DNA mutations per se being a primary driver of somatic stem cell aging.


Molecular and Cellular Biology | 2007

Critical Role for Ebf1 and Ebf2 in the Adipogenic Transcriptional Cascade

Maria Jimenez; Peter Åkerblad; Mikael Sigvardsson; Evan D. Rosen

ABSTRACT The Ebf (O/E) family of helix-loop-helix transcription factors plays a significant role in B lymphocyte and neuronal development. The three primary members of this family, Ebf1, 2, and 3, are all expressed in adipocytes, and Ebf1 promotes adipogenesis when overexpressed in NIH 3T3 fibroblasts. Here we report that these three proteins have adipogenic potential in multiple cellular models and that peroxisome proliferator-activated receptor γ (PPARγ) is required for this effect, at least in part due to direct activation of the PPARγ1 promoter by Ebf1. Ebf1 also directly binds to and activates the C/EBPα promoter, which exerts positive feedback on C/EBPδ expression. Despite this, C/EBPα is dispensable for the adipogenic action of Ebf proteins. Ebf1 itself is induced by C/EBPβ and δ, which bind and activate its promoter. Reduction of Ebf1 and Ebf2 proteins by specific short hairpin RNA blocks differentiation of 3T3-L1 cells, suggesting a critical role for these factors and the absence of functional redundancy between members of this family. Altogether, these data place Ebf1 within the known transcriptional cascade of adipogenesis and suggest critical roles for Ebf1 and Ebf2.


Journal of Immunology | 2008

EBF1 Is Essential for B-Lineage Priming and Establishment of a Transcription Factor Network in Common Lymphoid Progenitors

Sasan Zandi; Robert Månsson; Panagiotis Tsapogas; Jenny Zetterblad; David Bryder; Mikael Sigvardsson

Development of B-lymphoid cells in the bone marrow is a process under strict control of a hierarchy of transcription factors. To understand the development of a B-lymphoid-restricted functional network of transcription factors, we have investigated the cell autonomous role of the transcription factor EBF1 in early B cell development. This revealed that even though transplanted EBF1-deficient fetal liver cells were able to generate common lymphoid progenitors (CLPs) as well as B220+CD43+AA4.1+ candidate precursor B cells, none of these populations showed signs of B lineage priming. The isolated CLPs were able to generate T lymphocytes in vitro supporting the idea that the phenotype of EBF1-deficient mice is restricted to the development of the B lineage. Furthermore, EBF deficient CLPs displayed a reduction in Ig H chain recombination as compared with their wild-type counterpart and essentially lacked transcription of B-lineage-associated genes. Among the genes displaying reduced expression in the EBF1 deficient CLPs were the transcription factors Pax5, Pou2af1 (OcaB), and FoxO1 that all appear to be direct genetic targets for EBF1 because their promoters contained functional binding sites for this factor. This leads us to suggest that EBF1 regulates a transcription factor network crucial for B lineage commitment.

Collaboration


Dive into the Mikael Sigvardsson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Qian

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar

Sten Eirik W. Jacobsen

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge