Network


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

Hotspot


Dive into the research topics where Michael S. Krangel is active.

Publication


Featured researches published by Michael S. Krangel.


Nature Immunology | 2002

Regulation of the TCRalpha repertoire by the survival window of CD4(+)CD8(+) thymocytes.

Jian Guo; Abbas Hawwari; Hong Li; Zuoming Sun; Sanjeev K. Mahanta; Dan R. Littman; Michael S. Krangel; You-Wen He

T cell receptor (TCR) α alleles undergo primary and secondary rearrangement in double-positive (DP) thymocytes. By analyzing TCRα rearrangement in orphan nuclear receptor RORγ-deficient mice, in which the DP lifespan is shorter, and in Bcl-xL–transgenic mice, in which the DP lifespan is extended, we show that the progression of secondary Vα to Jα rearrangements is controlled by DP thymocyte survival. In addition, because Bcl-xL induces a bias towards 3′ Jα usage in peripheral T cells, we conclude that the programmed cell death of DP thymocytes is not simply a consequence of failed positive selection. Rather, it limits the progression of rearrangement along the Jα locus and the opportunities for positive selection, thereby regulating the TCRα repertoire.


Cell | 1979

Assembly and maturation of HLA-A and HLA-B antigens in vivo.

Michael S. Krangel; Harry T. Orr; Jack L. Strominger

HLA-A and HLA-B antigens are integral membrane glycoproteins which consist of a glycosylated heavy chain embedded in the membrane in noncovalent association with beta 2-microglobulin, a water-soluble polypeptide. The assembly and maturation of these antigens has been studied in vivo in the human B lymphoblastoid cell line T5-1 (HLA-A1, -A2, -B8, -B27). Two antigenically distinct populations of HLA-A and -B heavy chains can be detected by antisera which recognize determinants sensitive to the conformation of the heavy chain. One heavy chain population is associated with beta 2-microglobulin, whereas the other population is not. These populations can be further distinguished by their oligosaccharide structure and their localization within the cell. Pulse-chase experiments demonstrate a precursor-product relationship between these heavy chain populations and suggest the following pathway for the assembly and maturation of HLA-A and -B antigens. The completed heavy chains initially carry high mannose oligosaccharides and are largely or wholly associated with beta 2-microglobulin. During the next 10-15 min, association with beta 2-microglobulin occurs and the heavy chain conformation is altered. Beginning at about 30 min after synthesis, the oligosaccharides are converted from the high mannose form to the complex form, and between 60 and 80 min after synthesis, the mature antigens appear at the cell surface. These observations are discussed in relation to in vivo and in vitro studies on the biosynthesis of a variety of secreted proteins and membrane proteins.


Nature | 2011

A role for cohesin in T cell receptor rearrangement and thymocyte differentiation

Vlad C. Seitan; Bingtao Hao; Kikuë Tachibana-Konwalski; Thais Lavagnolli; Hegias Mira-Bontenbal; Karen E. Brown; Grace Teng; Tom Carroll; Anna Terry; Katie Horan; Hendrik Marks; David J. Adams; David G. Schatz; Luis Aragón; Amanda G. Fisher; Michael S. Krangel; Kim Nasmyth; Matthias Merkenschlager

Cohesin enables post-replicative DNA repair and chromosome segregation by holding sister chromatids together from the time of DNA replication in S phase until mitosis. There is growing evidence that cohesin also forms long-range chromosomal cis-interactions and may regulate gene expression in association with CTCF, mediator or tissue-specific transcription factors. Human cohesinopathies such as Cornelia de Lange syndrome are thought to result from impaired non-canonical cohesin functions, but a clear distinction between the cell-division-related and cell-division-independent functions of cohesion—as exemplified in Drosophila—has not been demonstrated in vertebrate systems. To address this, here we deleted the cohesin locus Rad21 in mouse thymocytes at a time in development when these cells stop cycling and rearrange their T-cell receptor (TCR) α locus (Tcra). Rad21-deficient thymocytes had a normal lifespan and retained the ability to differentiate, albeit with reduced efficiency. Loss of Rad21 led to defective chromatin architecture at the Tcra locus, where cohesion-binding sites flank the TEA promoter and the Eα enhancer, and demarcate Tcra from interspersed Tcrd elements and neighbouring housekeeping genes. Cohesin was required for long-range promoter–enhancer interactions, Tcra transcription, H3K4me3 histone modifications that recruit the recombination machinery and Tcra rearrangement. Provision of pre-rearranged TCR transgenes largely rescued thymocyte differentiation, demonstrating that among thousands of potential target genes across the genome, defective Tcra rearrangement was limiting for the differentiation of cohesin-deficient thymocytes. These findings firmly establish a cell-division-independent role for cohesin in Tcra locus rearrangement and provide a comprehensive account of the mechanisms by which cohesin enables cellular differentiation in a well-characterized mammalian system.


Molecular and Cellular Biology | 1994

Regulation of the T-cell receptor delta enhancer by functional cooperation between c-Myb and core-binding factors.

Cristina Hernández-Munain; Michael S. Krangel

A T-cell-specific transcriptional enhancer lies within the J delta 3-C delta intron of the human T-cell receptor (TCR) delta gene. The 30-bp minimal enhancer element denoted delta E3 carries a core sequence (TGTGGTTT) that binds a T-cell-specific factor, and that is necessary but not sufficient for transcriptional activation. Here we demonstrate that the transcription factor c-Myb regulates TCR delta enhancer activity through a binding site in delta E3 that is adjacent to the core site. Both v-Myb and c-Myb bind specifically to delta E3. The Myb site is necessary for enhancer activity, because a mutation that eliminates Myb binding abolishes transcriptional activation by the delta E3 element and by the 370-bp TCR delta enhancer. Transfection of cells with a c-Myb expression construct upregulates delta E3 enhancer activity, whereas treatment of cells with an antisense c-myb oligonucleotide inhibits delta E3 enhancer activity. Since intact Myb and core sites are both required for delta E3 function, our data argue that c-Myb and core binding factors must cooperate to mediate transcriptional activation through delta E3. Efficient cooperation depends on the relative positioning of the Myb and core sites, since only one of two overlapping Myb sites within delta E3 is functional and alterations of the distance between this site and the core site disrupt enhancer activity. Cooperative regulation by c-Myb and core-binding factors is likely to play an important role in the control of gene expression during T-cell development.


Nature Immunology | 2006

Regulation of T cell receptor-alpha gene recombination by transcription.

Iratxe Abarrategui; Michael S. Krangel

Despite the longstanding correlation between transcription and variable-(diversity)-joining (V(D)J) recombination, it is unknown whether transcription itself can direct recombinase targeting. Here we show that blockade of transcriptional elongation through the mouse T cell receptor-α (Tcra) locus suppressed Vα-to-Jα recombination and chromatin remodeling of Jα segments. Transcriptional blockade also derepressed cryptic Jα promoters. Our results demonstrate two key functions for transcription in Tcra locus regulation. Transcription increases the recombination of Jα segments located within several kilobases of a promoter and prevents the activation of downstream promoters through transcriptional interference. These influences promote an ordered progression of Tcra locus recombination events and selection of a robust Tcra repertoire.


Advances in Immunology | 1988

The γδT Cell Receptor

Michael B. Brenner; Jack L. Strominger; Michael S. Krangel

Publisher Summary Along with important conceptual similarities, there are also fundamental differences in structure and in the mode of operation of the antigen receptors on B and T cells. The B lymphocyte receptor is the immunoglobulin molecule. The large number of antibody molecules that can be produced by the organism result in part from somatic recombination of germ-line variable (V), diversity (D), joining (J), and constant (C) gene segments to form a contiguous (rearranged) gene, encoding an immunoglobulin polypeptide chain. Antibody diversity is extensive and is contributed by hundreds of V gene segments, dozens of D gene segments, and several J gene segments. The paired light and heavy chains of each antibody molecule form a unique site that is involved in the recognition of antigen; in turn, this unique site may be recognized by other antibodies as an idiotypic determinant. T cells differ from B cells in antigen recognition, because they often recognize different determinants, generally do not react with soluble or free antigen, and recognize antigen on the cell surface and only in conjunction with products encoded by self-major histocompatibility complex (MHC) genes. The 55 kDa species could be immunoprecipitated using antiserum made against synthetic peptides corresponding to the deduced amino acid sequence of a human TCR γ cDNA clone, suggesting that it was the elusive protein product of the TCR γ gene. The 40-kDa species failed to react with anti-TCR γ sera, and thus appeared to represent an additional component of the TCR γ –CD3 complex, which was termed “TCR δ .”


Current Opinion in Immunology | 2009

Mechanics of T cell receptor gene rearrangement

Michael S. Krangel

The four T cell receptor genes (Tcra, Tcrb, Tcrg, Tcrd) are assembled by V(D)J recombination according to distinct programs during intrathymic T cell development. These programs depend on genetic factors, including gene segment order and recombination signal sequences. They also depend on epigenetic factors. Regulated changes in chromatin structure, directed by enhancers and promoter, can modify the availability of recombination signal sequences to the RAG recombinase. Regulated changes in locus conformation may control the synapsis of distant recombination signal sequences, and regulated changes in subnuclear positioning may influence locus recombination events by unknown mechanisms. Together these influences may explain the ordered activation and inactivation of T cell receptor locus recombination events and the phenomenon of Tcrb allelic exclusion.


The EMBO Journal | 2007

Noncoding transcription controls downstream promoters to regulate T‐cell receptor α recombination

Iratxe Abarrategui; Michael S. Krangel

The T early α (TEA) promoter in the murine Tcra locus generates noncoding transcripts that extend across the 65 kb Jα array. Here, we have analyzed the significance of TEA transcription for Tcra locus regulation through the targeted introduction of a transcription terminator downstream of the TEA promoter. We demonstrate that noncoding transcription driven by this single promoter can instruct both positively and negatively the activity of downstream Jα promoters, and can similarly instruct alterations in Jα chromatin structure and Jα recombination. TEA transcription activates promoters associated with relatively proximal Jα segments and stimulates histone acetylation, histone methylation and chromatin accessibility in this region. In contrast, at more distal locations, TEA transcription inhibits promoter activity through transcriptional interference and suppresses chromatin accessibility. In combination, these effects target initial Vα‐to‐Jα recombination to TEA‐proximal Jα segments and promote the ordered usage of the Jα array. The ability of TEA transcription to coordinate the activity of multiple downstream promoters maximizes the biological potential of the Jα array and diversifies the Tcra repertoire.


Molecular and Cellular Biology | 1995

c-Myb and core-binding factor/PEBP2 display functional synergy but bind independently to adjacent sites in the T-cell receptor delta enhancer.

Cristina Hernández-Munain; Michael S. Krangel

A T-cell-specific transcriptional enhancer lies within the J delta 3-C delta intron of the human T-cell receptor delta gene. We have previously shown that a 30-bp element, denoted delta E3, acts as the minimal TCR delta enhancer and that within delta E3, adjacent and precisely spaced binding sites for core-binding factor (CBF/PEBP2) and c-Myb are essential for transcriptional activity. These data suggested that CBF/PEBP2 and c-Myb synergize to mediate transcriptional activity but did not establish the molecular basis for synergy. In this study, we have examined in detail the binding of CBF/PEBP2 and c-Myb to delta E3. We found that CBF/PEBP2 and c-Myb could simultaneously occupy the core site and one of two overlapping Myb sites within delta E3. However, equilibrium binding and kinetic dissociation experiments suggest that the two factors bind to delta E3 independently, rather than cooperatively. This was found to be true by using isoforms of these factors present in extracts of transfected COS-7 cells, as well as the natural factors present in nuclear extracts of the Jurkat T-cell line. We further showed that CBF/PEBP2 and c-Myb provide unique transactivation functions, since the core-Myb combination cannot be substituted by dimerized core or Myb sites. We propose that spatially precise synergy between CBF/PEBP2 and c-Myb may result from the ability of the two factors to form a composite surface that makes unique and stereospecific contacts with one or more additional components of the transcriptional machinery.


Nature Immunology | 2004

Targeted inhibition of V(D)J recombination by a histone methyltransferase.

Oleg Osipovich; Robin Milley; Amber Meade; Makoto Tachibana; Yoichi Shinkai; Michael S. Krangel; Eugene M. Oltz

The tissue- and stage-specific assembly of antigen receptor genes by V(D)J recombination is regulated by changes in the chromatin accessibility of target gene segments. This dynamic remodeling process is coordinated by cis-acting promoters and enhancers, which function as accessibility control elements. The basic epigenetic mechanisms that activate or repress chromatin accessibility to V(D)J recombinase remain unclear. We now demonstrate that a histone methyltransferase overrides accessibility control element function and cripples V(D)J recombination of chromosomal gene segments. The recruited histone methyltransferase induces extensive revisions in the local chromatin environment, including altered histone modifications and de novo methylation of DNA. These findings indicate a key function for histone methyltransferases in the tissue- and stage-specific suppression of antigen receptor gene assembly during lymphocyte development.

Collaboration


Dive into the Michael S. Krangel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael B. Brenner

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cristina Hernández-Munain

Spanish National Research Council

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