Network


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

Hotspot


Dive into the research topics where Jeffrey S. Haug is active.

Publication


Featured researches published by Jeffrey S. Haug.


Nature | 2006

PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention

Jiwang Zhang; Justin C. Grindley; Tong Yin; Sachintha Jayasinghe; Xi C. He; Jason T. Ross; Jeffrey S. Haug; Dawn Rupp; Kimberly S. Porter-Westpfahl; Leanne M. Wiedemann; Hong Wu; Linheng Li

Haematopoietic stem cells (HSCs) must achieve a balance between quiescence and activation that fulfils immediate demands for haematopoiesis without compromising long-term stem cell maintenance, yet little is known about the molecular events governing this balance. Phosphatase and tensin homologue (PTEN) functions as a negative regulator of the phosphatidylinositol-3-OH kinase (PI(3)K)–Akt pathway, which has crucial roles in cell proliferation, survival, differentiation and migration. Here we show that inactivation of PTEN in bone marrow HSCs causes their short-term expansion, but long-term decline, primarily owing to an enhanced level of HSC activation. PTEN-deficient HSCs engraft normally in recipient mice, but have an impaired ability to sustain haematopoietic reconstitution, reflecting the dysregulation of their cell cycle and decreased retention in the bone marrow niche. Mice with PTEN-mutant bone marrow also have an increased representation of myeloid and T-lymphoid lineages and develop myeloproliferative disorder (MPD). Notably, the cell populations that expand in PTEN mutants match those that become dominant in the acute myeloid/lymphoid leukaemia that develops in the later stages of MPD. Thus, PTEN has essential roles in restricting the activation of HSCs, in lineage fate determination, and in the prevention of leukaemogenesis.


Cell | 2012

Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche

Ryohichi Sugimura; Xi C. He; Aparna Venkatraman; Fumio Arai; Andrew C. Box; Craig L. Semerad; Jeffrey S. Haug; Lai Peng; Xiao-bo Zhong; Toshio Suda; Linheng Li

Wnt signaling is involved in self-renewal and maintenance of hematopoietic stem cells (HSCs); however, the particular role of noncanonical Wnt signaling in regulating HSCs in vivo is largely unknown. Here, we show Flamingo (Fmi) and Frizzled (Fz) 8, members of noncanonical Wnt signaling, both express in and functionally maintain quiescent long-term HSCs. Fmi regulates Fz8 distribution at the interface between HSCs and N-cadherin(+) osteoblasts (N-cad(+)OBs that enrich osteoprogenitors) in the niche. We further found that N-cad(+)OBs predominantly express noncanonical Wnt ligands and inhibitors of canonical Wnt signaling under homeostasis. Under stress, noncanonical Wnt signaling is attenuated and canonical Wnt signaling is enhanced in activation of HSCs. Mechanistically, noncanonical Wnt signaling mediated by Fz8 suppresses the Ca(2+)-NFAT- IFNγ pathway, directly or indirectly through the CDC42-CK1α complex and also antagonizes canonical Wnt signaling in HSCs. Taken together, our findings demonstrate that noncanonical Wnt signaling maintains quiescent long-term HSCs through Fmi and Fz8 interaction in the niche.


Nature | 2013

Maternal imprinting at the H19–Igf2 locus maintains adult haematopoietic stem cell quiescence

Aparna Venkatraman; Xi C. He; Joanne L. Thorvaldsen; Ryohichi Sugimura; John M. Perry; Fang Tao; Meng Zhao; Matthew K. Christenson; Rebeca Sanchez; Jaclyn Y. Yu; Lai Peng; Jeffrey S. Haug; Ariel Paulson; Hua Li; Xiao-bo Zhong; Thomas L. Clemens; Marisa S. Bartolomei; Linheng Li

The epigenetic regulation of imprinted genes by monoallelic DNA methylation of either maternal or paternal alleles is critical for embryonic growth and development. Imprinted genes were recently shown to be expressed in mammalian adult stem cells to support self-renewal of neural and lung stem cells; however, a role for imprinting per se in adult stem cells remains elusive. Here we show upregulation of growth-restricting imprinted genes, including in the H19–Igf2 locus, in long-term haematopoietic stem cells and their downregulation upon haematopoietic stem cell activation and proliferation. A differentially methylated region upstream of H19 (H19-DMR), serving as the imprinting control region, determines the reciprocal expression of H19 from the maternal allele and Igf2 from the paternal allele. In addition, H19 serves as a source of miR-675, which restricts Igf1r expression. We demonstrate that conditional deletion of the maternal but not the paternal H19-DMR reduces adult haematopoietic stem cell quiescence, a state required for long-term maintenance of haematopoietic stem cells, and compromises haematopoietic stem cell function. Maternal-specific H19-DMR deletion results in activation of the Igf2–Igfr1 pathway, as shown by the translocation of phosphorylated FoxO3 (an inactive form) from nucleus to cytoplasm and the release of FoxO3-mediated cell cycle arrest, thus leading to increased activation, proliferation and eventual exhaustion of haematopoietic stem cells. Mechanistically, maternal-specific H19-DMR deletion leads to Igf2 upregulation and increased translation of Igf1r, which is normally suppressed by H19-derived miR-675. Similarly, genetic inactivation of Igf1r partly rescues the H19-DMR deletion phenotype. Our work establishes a new role for this unique form of epigenetic control at the H19–Igf2 locus in maintaining adult stem cells.


Cell Stem Cell | 2008

N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells.

Jeffrey S. Haug; Xi C. He; Justin C. Grindley; Joshua P. Wunderlich; Karin Gaudenz; Jason T. Ross; Ariel Paulson; Kathryn P. Wagner; Yucai Xie; Ruihong Zhu; Tong Yin; John M. Perry; Mark J. Hembree; Erin P. Redenbaugh; Glenn L. Radice; Christopher Seidel; Linheng Li

Osteoblasts expressing the homophilic adhesion molecule N-cadherin form a hematopoietic stem cell (HSC) niche. Therefore, we examined how N-cadherin expression in HSCs relates to their function. We found that bone marrow (BM) cells highly expressing N-cadherin (N-cadherin(hi)) are not stem cells, being largely devoid of a Lineage(-)Sca1(+)cKit(+) population and unable to reconstitute hematopoietic lineages in irradiated recipient mice. Instead, long-term HSCs form distinct populations expressing N-cadherin at intermediate (N-cadherin(int)) or low (N-cadherin(lo)) levels. The minority N-cadherin(lo) population can robustly reconstitute the hematopoietic system, express genes that may prime them to mobilize, and predominate among HSCs mobilized from BM to spleen. The larger N-cadherin(int) population performs poorly in reconstitution assays when freshly isolated but improves in response to overnight in vitro culture. Their expression profile and lower cell-cycle entry rate suggest N-cadherin(int) cells are being held in reserve. Thus, differential N-cadherin expression reflects functional distinctions between two HSC subpopulations.


Gastroenterology | 2013

Isolation and Characterization of Intestinal Stem Cells Based on Surface Marker Combinations and Colony-Formation Assay

Fengchao Wang; David Scoville; Xi C. He; Maxime M. Mahe; Andrew C. Box; John M. Perry; Nicholas R. Smith; Nan Ye Lei; Paige S. Davies; Megan K. Fuller; Jeffrey S. Haug; Melainia McClain; Adam D. Gracz; Sheng Ding; Matthias Stelzner; James C.Y. Dunn; Scott T. Magness; Melissa H. Wong; Martin G. Martin; Michael A. Helmrath; Linheng Li

BACKGROUND & AIMS Identification of intestinal stem cells (ISCs) has relied heavily on the use of transgenic reporters in mice, but this approach is limited by mosaic expression patterns and difficult to directly apply to human tissues. We sought to identify reliable surface markers of ISCs and establish a robust functional assay to characterize ISCs from mouse and human tissues. METHODS We used immunohistochemistry, real-time reverse-transcription polymerase chain reaction, and fluorescence-activated cell sorting (FACS) to analyze intestinal epithelial cells isolated from mouse and human intestinal tissues. We compared different combinations of surface markers among ISCs isolated based on expression of Lgr5-green fluorescent protein. We developed a culture protocol to facilitate the identification of functional ISCs from mice and then tested the assay with human intestinal crypts and putative ISCs. RESULTS CD44(+)CD24(lo)CD166(+) cells, isolated by FACS from mouse small intestine and colon, expressed high levels of stem cell-associated genes. Transit-amplifying cells and progenitor cells were then excluded based on expression of GRP78 or c-Kit. CD44(+)CD24(lo)CD166(+) GRP78(lo/-) putative stem cells from mouse small intestine included Lgr5-GFP(hi) and Lgr5-GFP(med/lo) cells. Incubation of these cells with the GSK inhibitor CHIR99021 and the E-cadherin stabilizer Thiazovivin resulted in colony formation by 25% to 30% of single-sorted ISCs. CONCLUSIONS We developed a culture protocol to identify putative ISCs from mouse and human tissues based on cell surface markers. CD44(+)CD24(lo)CD166(+), GRP78(lo/-), and c-Kit(-) facilitated identification of putative stem cells from the mouse small intestine and colon, respectively. CD44(+)CD24(-/lo)CD166(+) also identified putative human ISCs. These findings will facilitate functional studies of mouse and human ISCs.


Genes & Development | 2011

Cooperation between both Wnt/{beta}-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion.

John M. Perry; Xi C. He; Ryohichi Sugimura; Justin C. Grindley; Jeffrey S. Haug; Sheng Ding; Linheng Li

Although self-renewal is the central property of stem cells, the underlying mechanism remains inadequately defined. Using a hematopoietic stem and progenitor cell (HSPC)-specific conditional induction line, we generated a compound genetic model bearing both Pten deletion and β-catenin activation. These double mutant mice exhibit a novel phenotype, including expansion of phenotypic long-term hematopoietic stem cells (LT-HSCs) without extensive differentiation. Unexpectedly, constitutive activation of β-catenin alone results in apoptosis of HSCs. However, together, the Wnt/β-catenin and PTEN/PI3k/Akt pathways interact to drive phenotypic LT-HSC expansion by inducing proliferation while simultaneously inhibiting apoptosis and blocking differentiation, demonstrating the necessity of complementary cooperation between the two pathways in promoting self-renewal. Mechanistically, β-catenin activation reduces multiple differentiation-inducing transcription factors, blocking differentiation partially through up-regulation of Inhibitor of differentiation 2 (Id2). In double mutants, loss of Pten enhances the HSC anti-apoptotic factor Mcl-1. All of these contribute in a complementary way to HSC self-renewal and expansion. While permanent, genetic alteration of both pathways in double mutant mice leads to expansion of phenotypic HSCs, these HSCs cannot function due to blocked differentiation. We developed a pharmacological approach to expand normal, functional HSCs in culture using factors that reversibly activate both Wnt/β-catenin and PI3K/Akt signaling simultaneously. We show for the first time that activation of either single pathway is insufficient to expand primitive HSCs, but in combination, both pathways drive self-renewal and expansion of HSCs with long-term functional capacity.


Cell | 2009

DNA-PKcs-PIDDosome: a nuclear caspase-2-activating complex with role in G2/M checkpoint maintenance.

Mingan Shi; Carolyn J. Vivian; Kyung Jong Lee; Chunmin Ge; Keiko Morotomi-Yano; Claudia Manzl; Florian J. Bock; Shigeo Sato; Chieri Tomomori-Sato; Ruihong Zhu; Jeffrey S. Haug; Selene K. Swanson; Michael P. Washburn; David J. Chen; Benjamin P C Chen; Andreas Villunger; Laurence Florens; Chunying Du

A reciprocating piston type compressor having a cylinder block, a plurality of cylinder bores, and at least a housing closing an end of the cylinder block. The housing contains a suction chamber for a refrigerant gas to be compressed and a discharge chamber for the compressed refrigerant gas discharged from the cylinder bores in response to reciprocation of a plurality of pistons. The compressed gas is discharged through discharge ports closed by a discharge valve element having a plurality of integral discharge reed-valves movable between a closed positions and a predetermined open positions. The open position is defined by a stop unit integrally formed in an inner wall of the housing. The stop unit has a plurality of flat stop faces formed on the inner wall to permit free ends of the discharge reed-valves to come into contact engagement therewith, when the discharge reed-valves are moved from the closed positions to the open positions.Caspase-2 is unique among all the mammalian caspases in that it is the only caspase that is present constitutively in the cell nucleus, in addition to other cellular compartments. However, the functional significance of this nuclear localization is unknown. Here we show that DNA damage induced by gamma-radiation triggers the phosphorylation of nuclear caspase-2 at the S122 site within its prodomain, leading to its cleavage and activation. This phosphorylation is carried out by the nuclear serine/threonine protein kinase DNA-PKcs and promoted by the p53-inducible death-domain-containing protein PIDD within a large nuclear protein complex consisting of DNA-PKcs, PIDD, and caspase-2, which we have named the DNA-PKcs-PIDDosome. This phosphorylation and the catalytic activity of caspase-2 are involved in the maintenance of a G2/M DNA damage checkpoint and DNA repair mediated by the nonhomologous end-joining (NHEJ) pathway. The DNA-PKcs-PIDDosome thus represents a protein complex that impacts mammalian G2/M DNA damage checkpoint and NHEJ.


Cell Stem Cell | 2016

The Dlk1-Gtl2 Locus Preserves LT-HSC Function by Inhibiting the PI3K-mTOR Pathway to Restrict Mitochondrial Metabolism

Pengxu Qian; Xi C. He; Ariel Paulson; Zhenrui Li; Fang Tao; John M. Perry; Fengli Guo; Meng Zhao; Lei Zhi; Aparna Venkatraman; Jeffrey S. Haug; Tari Parmely; Hua Li; Rick T. Dobrowsky; Weng-Xing Ding; Tomohiro Kono; Anne C. Ferguson-Smith; Linheng Li

The mammalian imprinted Dlk1-Gtl2 locus produces multiple non-coding RNAs (ncRNAs) from the maternally inherited allele, including the largest miRNA cluster in the mammalian genome. This locus has characterized functions in some types of stem cell, but its role in hematopoietic stem cells (HSCs) is unknown. Here, we show that the Dlk1-Gtl2 locus plays a critical role in preserving long-term repopulating HSCs (LT-HSCs). Through transcriptome profiling in 17 hematopoietic cell types, we found that ncRNAs expressed from the Dlk1-Gtl2 locus are predominantly enriched in fetal liver HSCs and the adult LT-HSC population and sustain long-term HSC functionality. Mechanistically, the miRNA mega-cluster within the Dlk1-Gtl2 locus suppresses the entire PI3K-mTOR pathway. This regulation in turn inhibits mitochondrial biogenesis and metabolic activity and protects LT-HSCs from excessive reactive oxygen species (ROS) production. Our data therefore show that the imprinted Dlk1-Gtl2 locus preserves LT-HSC function by restricting mitochondrial metabolism.


Molecular and Cellular Biology | 2012

Polycomb Repressive Complex 2-Dependent and -Independent Functions of Jarid2 in Transcriptional Regulation in Drosophila

Hans Martin Herz; Man Mohan; Alexander S. Garrett; Caitlynn Miller; David Casto; Ying Zhang; Christopher Seidel; Jeffrey S. Haug; Laurence Florens; Michael P. Washburn; Masamitsu Yamaguchi; Ramin Shiekhattar; Ali Shilatifard

ABSTRACT Jarid2 was recently identified as an important component of the mammalian Polycomb repressive complex 2 (PRC2), where it has a major effect on PRC2 recruitment in mouse embryonic stem cells. Although Jarid2 is conserved in Drosophila, it has not previously been implicated in Polycomb (Pc) regulation. Therefore, we purified Drosophila Jarid2 and its associated proteins and found that Jarid2 associates with all of the known canonical PRC2 components, demonstrating a conserved physical interaction with PRC2 in flies and mammals. Furthermore, in vivo studies with Jarid2 mutants in flies demonstrate that among several histone modifications tested, only methylation of histone 3 at K27 (H3K27), the mark implemented by PRC2, was affected. Genome-wide profiling of Jarid2, Su(z)12 (Suppressor of zeste 12), and H3K27me3 occupancy by chromatin immunoprecipitation with sequencing (ChIP-seq) indicates that Jarid2 and Su(z)12 have very similar distribution patterns on chromatin. However, Jarid2 and Su(z)12 occupancy levels at some genes are significantly different, with Jarid2 being present at relatively low levels at many Pc response elements (PREs) of certain Homeobox (Hox) genes, providing a rationale for why Jarid2 was never identified in Pc screens. Gene expression analyses show that Jarid2 and E(z) (Enhancer of zeste, a canonical PRC2 component) are not only required for transcriptional repression but might also function in active transcription. Identification of Jarid2 as a conserved PRC2 interactor in flies provides an opportunity to begin to probe some of its novel functions in Drosophila development.


Blood | 2012

FGF signaling facilitates postinjury recovery of mouse hematopoietic system.

Meng Zhao; Jason T. Ross; Tomer Itkin; John M. Perry; Aparna Venkatraman; Jeffrey S. Haug; Mark J. Hembree; Chuxia Deng; Tsvee Lapidot; Xi C. He; Linheng Li

Previous studies have shown that fibroblast growth factor (FGF) signaling promotes hematopoietic stem and progenitor cell (HSPC) expansion in vitro. However, it is unknown whether FGF promotes HSPC expansion in vivo. Here we examined FGF receptor 1 (FGFR1) expression and investigated its in vivo function in HSPCs. Conditional knockout (CKO) of Fgfr1 did not affect phenotypical number of HSPCs and homeostatic hematopoiesis, but led to a reduced engraftment only in the secondary transplantation. When treated with 5-fluorouracil (5FU), the Fgfr1 CKO mice showed defects in both proliferation and subsequent mobilization of HSPCs. We identified megakaryocytes (Mks) as a major resource for FGF production, and further discovered a novel mechanism by which Mks underwent FGF-FGFR signaling dependent expansion to accelerate rapid FGF production under stress. Within HSPCs, we observed an up-regulation of nuclear factor κB and CXCR4, a receptor for the chemoattractant SDF-1, in response to bone marrow damage only in control but not in Fgfr1 CKO model, accounting for the corresponding defects in proliferation and migration of HSPCs. This study provides the first in vivo evidence that FGF signaling facilitates postinjury recovery of the mouse hematopoietic system by promoting proliferation and facilitating mobilization of HSPCs.

Collaboration


Dive into the Jeffrey S. Haug's collaboration.

Top Co-Authors

Avatar

Xi C. He

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Linheng Li

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

John M. Perry

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Andrew C. Box

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ariel Paulson

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Fang Tao

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Hua Li

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Laurence Florens

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Michael P. Washburn

Stowers Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Ruihong Zhu

Stowers Institute for Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge