551
|
Neutrophil-specific deletion of Syk kinase results in reduced host defense to bacterial infection. Blood 2009; 114:4871-82. [PMID: 19797524 DOI: 10.1182/blood-2009-05-220806] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukocyte-specific CD18 integrins are critical in mediating cell recruitment and activation during host defense responses to bacterial infection. The signaling pathways downstream of CD18 integrins are dependent on the spleen tyrosine kinase, Syk. To investigate the role integrin signaling plays in host defense, we examined the responses of Syk-deficient neutrophils to bacterial challenge with serum-opsonized Staphylococcus aureus and Escherichia coli. Syk-conditional knockout mice lacking this kinase specifically in myeloid cells or just neutrophils were also used to investigate host responses in vivo. Syk-deficient neutrophils manifested impaired exocytosis of secondary and tertiary granules, reduced cytokine release, and very poor activation of the NADPH oxidase in response to serum-opsonized S aureus and E coli. These functional defects correlated with impaired activation of c-Cbl, Pyk2, Erk1/2, and p38 kinases. Bacterial phagocytosis, neutrophil extracellular trap formation, and killing were also reduced in Syk-deficient cells, with a more profound effect after S aureus challenge. In vivo, loss of Syk in myeloid cells or specifically in neutrophils resulted in reduced clearance of S aureus after subcutaneous or intraperitoneal infection, despite normal recruitment of inflammatory cells. These results indicate that loss of Syk kinase-mediated integrin signaling impairs leukocyte activation, leading to reduced host defense responses.
Collapse
|
552
|
Fedeli M, Napolitano A, Wong MPM, Marcais A, de Lalla C, Colucci F, Merkenschlager M, Dellabona P, Casorati G. Dicer-dependent microRNA pathway controls invariant NKT cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:2506-12. [PMID: 19625646 DOI: 10.4049/jimmunol.0901361] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Invariant NK T (iNKT) cells are a separate lineage of T lymphocytes with innate effector functions. They express an invariant TCR specific for lipids presented by CD1d and their development and effector differentiation rely on a unique gene expression program. We asked whether this program includes microRNAs, small noncoding RNAs that regulate gene expression posttranscriptionally and play a key role in the control of cellular differentiation programs. To this aim, we investigated iNKT cell development in mice in which Dicer, the RNase III enzyme that generates functional microRNAs, is deleted in cortical thymocytes. We find that Dicer deletion results in a substantial reduction of iNKT cells in thymus and their disappearance from the periphery, unlike mainstream T cells. Without Dicer, iNKT cells do not complete their innate effector differentiation and display a defective homeostasis due to increased cell death. Differentiation and homeostasis of iNKT cells require Dicer in a cell-autonomous fashion. Furthermore, we identify a miRNA profile specific for iNKT cells, which exhibits features of activated/effector T lymphocytes, consistent with the idea that iNKT cells undergo agonist thymic selection. Together, these results define a critical role of the Dicer-dependent miRNA pathway in the physiology of iNKT cells.
Collapse
Affiliation(s)
- Maya Fedeli
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, H San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
553
|
Dumont C, Corsoni-Tadrzak A, Ruf S, de Boer J, Williams A, Turner M, Kioussis D, Tybulewicz VLJ. Rac GTPases play critical roles in early T-cell development. Blood 2009; 113:3990-8. [PMID: 19088377 PMCID: PMC2673125 DOI: 10.1182/blood-2008-09-181180] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 12/14/2008] [Indexed: 01/11/2023] Open
Abstract
The Rac1 and Rac2 GTPases play important roles in many processes including cytoskeletal reorganization, proliferation, and survival, and are required for B-cell development. Previous studies had shown that deficiency in Rac2 did not affect T-cell development, whereas the function of Rac1 in this process has not been investigated. We now show that simultaneous absence of both GTPases resulted in a very strong developmental block at the pre-TCR checkpoint and in defective positive selection. Unexpectedly, deficiency of Rac1 and Rac2 also resulted in the aberrant survival of thymocytes lacking expression of TCR beta, showing hallmarks of hyperactive Notch signaling. Furthermore, we found a similar novel phenotype in the absence of Vav1, Vav2, and Vav3, which function as guanine nucleotide exchange factors for Rac1 and Rac2. These results show that a pathway containing Vav and Rac proteins may negatively regulate Notch signaling during early thymic development.
Collapse
Affiliation(s)
- Celine Dumont
- Division of Immune Cell Biology, Medical Research Council (MRC) National Institute for Medical Research, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
554
|
Fusion of hematopoietic cells with Purkinje neurons does not lead to stable heterokaryon formation under noninvasive conditions. J Neurosci 2009; 29:3799-807. [PMID: 19321776 DOI: 10.1523/jneurosci.5848-08.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Transplanted hematopoietic cells have previously been shown to contribute to cells of other tissues by cell fusion. We wanted to elucidate whether this phenomenon of cell fusion also occurs under physiological conditions. Using a transgenic mouse reporter system to irreversibly label cells of the hematopoietic lineage, we were able to test their contribution to other tissues in the absence of any additional and potentially confounding factors such as irradiation or chemoablation. We found genetically marked, fused Purkinje neurons as well as hepatocytes in numbers comparable to previous bone marrow transplantation studies. The number of fused Purkinje neurons increased after intrathecal administration of bacterial lipopolysaccharide, suggesting that cell fusion can be induced by inflammation. In contrast to previous studies, however, genetically labeled Purkinje neurons never contained more than one nucleus, and we found only a single cell containing two Y-chromosomes in a male mouse. Consistent with results from the mouse model and unlike human bone marrow transplant recipients, postmortem adult human cerebelli of nontransplanted individuals were devoid of binucleated or polyploid Purkinje neurons. Therefore, our data suggests that fusion of hematopoietic cells with Purkinje neurons is only transient and does not lead to stable heterokaryon formation under noninvasive conditions.
Collapse
|
555
|
Scobie L, Hector RD, Grant L, Bell M, Nielsen AA, Meikle S, Philbey A, Philbey A, Thrasher AJ, Thrasher AJ, Cameron ER, Blyth K, Neil JC. A novel model of SCID-X1 reconstitution reveals predisposition to retrovirus-induced lymphoma but no evidence of gammaC gene oncogenicity. Mol Ther 2009; 17:1031-8. [PMID: 19337236 DOI: 10.1038/mt.2009.59] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The emergence of leukemia following gene transfer to restore common cytokine receptor gamma chain (gammaC) function in X-linked severe combined immunodeficiency (SCID-X1) has raised important questions with respect to gene therapy safety. To explore the risk factors involved, we tested the oncogenic potential of human gammaC in new strains of transgenic mice expressing the gene under the control of the CD2 promoter and locus control region (LCR). These mice demonstrated mildly perturbed T-cell development, with an increased proportion of thymic CD8 cells, but showed no predisposition to tumor development even on highly tumor prone backgrounds or after gamma-retrovirus infection. The human CD2-gammaC transgene rescued T and B-cell development in gammaC(-/-) mice but with an age-related delay, mimicking postnatal reconstitution in SCID-X1 gene therapy subjects. However, we noted that gammaC(-/-) mice are acutely susceptible to murine leukemia virus (MLV) leukemogenesis, and that this trait was not corrected by the gammaC transgene. We conclude that the SCID-X1 phenotype can be corrected safely by stable ectopic expression of gammaC and that the transgene is not significantly oncogenic when expressed in this context. However, an underlying predisposition conferred by the SCID-X1 background appears to collaborate with insertional mutagenesis to increase the risk of tumor development.
Collapse
Affiliation(s)
- Linda Scobie
- Division of Pathological Sciences, Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Glasgow, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
556
|
Outram SV, Hager-Theodorides AL, Shah DK, Rowbotham NJ, Drakopoulou E, Ross SE, Lanske B, Dessens JT, Crompton T. Indian hedgehog (Ihh) both promotes and restricts thymocyte differentiation. Blood 2009; 113:2217-28. [PMID: 19109233 DOI: 10.1182/blood-2008-03-144840] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We show that Indian Hedgehog (Ihh) regulates T-cell development and homeostasis in both fetal and adult thymus, controlling thymocyte number. Fetal Ihh(-/-) thymi had reduced differentiation to double-positive (DP) cell and reduced cell numbers compared with wild-type littermates. Surprisingly, fetal Ihh(+/-) thymi had increased thymocyte numbers and proportion of DP cells relative to wild type, indicating that Ihh also negatively regulates thymocyte development. In vitro treatment of thymus explants with exogenous recombinant Hedgehog protein promoted thymocyte development in Ihh(-/-) thymi but inhibited thymocyte development in Ihh(+/-), confirming both positive and negative regulatory functions of Ihh. Analysis of Rag(-/-)Ihh(+/-) thymi showed that Ihh promotes T-cell development before pre-T-cell receptor (pre-TCR) signaling, but negatively regulates T-cell development only after pre-TCR signaling has taken place. We show that Ihh is most highly expressed by the DP population and that Ihh produced by DP cells feeds back to negatively regulate the differentiation and proliferation of their double-negative progenitors. Thus, differentiation from double-negative to DP cell, and hence the size of the DP population, is dependent on the concentration of Ihh in the thymus. Analysis of Ihh conditional knockout and heterozygote adult mice showed that Ihh also influences thymocyte number in the adult.
Collapse
Affiliation(s)
- Susan V Outram
- Immunobiology Unit, University College London Institute of Child Health, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
557
|
Rongvaux A, Galli M, Denanglaire S, Van Gool F, Drèze PL, Szpirer C, Bureau F, Andris F, Leo O. Nicotinamide phosphoribosyl transferase/pre-B cell colony-enhancing factor/visfatin is required for lymphocyte development and cellular resistance to genotoxic stress. THE JOURNAL OF IMMUNOLOGY 2008; 181:4685-95. [PMID: 18802071 DOI: 10.4049/jimmunol.181.7.4685] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nicotinamide phosphoribosyl transferase (Nampt)/pre-B cell colony-enhancing factor (PBEF)/visfatin is a protein displaying multiple functional properties. Originally described as a cytokine-like protein able to regulate B cell development, apoptosis, and glucose metabolism, this protein also plays an important role in NAD biosynthesis. To gain insight into its physiological role, we have generated a mouse strain expressing a conditional Nampt allele. Lack of Nampt expression strongly affects development of both T and B lymphocytes. Analysis of hemizygous cells and in vitro cell lines expressing distinct levels of Nampt illustrates the critical role of this protein in regulating intracellular NAD levels. Consequently, a clear relationship was found between intracellular Nampt levels and cell death in response to the genotoxic agent MNNG (N-methyl-N'-nitro-N-nitrosoguanidine), confirming that this enzyme represents a key regulator of cell sensitivity to NAD-consuming stress secondary to poly(ADP-ribose) polymerases overactivation. By using mutant forms of this protein and a well-characterized pharmacological inhibitor (FK866), we unequivocally demonstrate that the ability of the Nampt to regulate cell viability during genotoxic stress requires its enzymatic activity. Collectively, these data demonstrate that Nampt participates in cellular resistance to genotoxic/oxidative stress, and it may confer to cells of the immune system the ability to survive during stressful situations such as inflammation.
Collapse
Affiliation(s)
- Anthony Rongvaux
- Laboratoire de Physiologie Animale, Institut de Biologie et Médecine Moléculaire (IBMM), Université Libre de Bruxelles, Gosselies, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
558
|
ORTOLANO F, MAFFIA P, DEVER G, HUTCHISON S, BENSON R, MILLINGTON OR, DE SIMONI MG, BUSHELL TJ, GARSIDE P, CARSWELL HV, BREWER JM. Imaging T-cell movement in the brain during experimental cerebral malaria. Parasite Immunol 2008; 31:147-50. [DOI: 10.1111/j.1365-3024.2008.01090.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
559
|
Moncrieffe H, Coles M, Stockinger B. The influence of CD4 T-cell subsets on control of CD4 T-cell-mediated graft-versus-host disease. Immunology 2008; 125:459-68. [PMID: 18498346 PMCID: PMC2612549 DOI: 10.1111/j.1365-2567.2008.02866.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 03/31/2008] [Accepted: 04/21/2008] [Indexed: 11/30/2022] Open
Abstract
In this study, we tested the effect of different T-cell subpopulations on antigen driven effector cell expansion in lymphopenic hosts, making use of an experimental model of graft-versus-host disease (GVHD). Fluorescence-activated cell sorted (FACS) naïve CD4 T cells from C57BL/6 mice, transferred into lymphopenic F1 (C57BL/6 x BALB/c) Rag-deficient hosts, proliferated extensively and migrated systemically causing acute GVHD within 4 weeks after transfer. Adoptive hosts of CD4 memory T cells on the other hand developed milder symptoms of GVHD with later onset. T-cell expansion and migration to peripheral sites as well as development of GVHD were prevented when naïve T cells were transferred together with CD4(+) CD25(+) T cells, but co-transfer of memory T cells with naïve T cells could not prevent GVHD, although its onset was delayed. OX40, a costimulatory marker that is upregulated at an early time point after T-cell activation and enhances T-cell proliferation, cytokine secretion and survival, was strongly upregulated during GVH responses. Naïve T cells deficient in OX40 expression caused markedly reduced GVH in onset and severity despite some level of expansion in the adoptive host, suggesting an important role of this molecule in the immune pathology of GVHD.
Collapse
Affiliation(s)
- Halima Moncrieffe
- Division of Molecular Immunology, The MRC National Institute for Medical Research, Mill Hill, London, UK.
| | | | | |
Collapse
|
560
|
Papapetrou EP, Kovalovsky D, Beloeil L, Sant'angelo D, Sadelain M. Harnessing endogenous miR-181a to segregate transgenic antigen receptor expression in developing versus post-thymic T cells in murine hematopoietic chimeras. J Clin Invest 2008; 119:157-68. [PMID: 19033646 DOI: 10.1172/jci37216] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 10/15/2008] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression by targeting complementary sequences, referred to as miRNA recognition elements (MREs), typically located in the 3' untranslated region of mRNAs. miR-181a is highly expressed in developing thymocytes and markedly downregulated in post-thymic T cells. We investigated whether endogenous miR-181a can be harnessed to segregate expression of chimeric antigen receptors (CARs) and TCRs between developing and mature T cells. Lentiviral-encoded antigen receptors were tagged with a miR-181a-specific MRE and transduced into mouse BM cells that were used to generate hematopoietic chimeras. Expression of a CAR specific for human CD19 (hCD19) was selectively suppressed in late double-negative and double-positive thymocytes, coinciding with the peak in endogenous miR-181a expression. Receptor expression was fully restored in post-thymic resting and activated T cells, affording protection against a subsequent challenge with hCD19+ tumors. Hematopoietic mouse chimeras engrafted with a conalbumin-specific TCR prone to thymic clonal deletion acquired peptide-specific T cell responsiveness only when the vector-encoded TCR transcript was similarly engineered to be subject to regulation by miR-181a. These results demonstrate the potential of miRNA-regulated transgene expression in stem cell-based therapies, including cancer immunotherapy.
Collapse
Affiliation(s)
- Eirini P Papapetrou
- Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
561
|
Dauner JG, Chappell CP, Williams IR, Jacob J. Perfusion fixation preserves enhanced yellow fluorescent protein and other cellular markers in lymphoid tissues. J Immunol Methods 2008; 340:116-22. [PMID: 19007785 DOI: 10.1016/j.jim.2008.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 10/13/2008] [Accepted: 10/15/2008] [Indexed: 12/21/2022]
Abstract
Fluorescent proteins are increasingly being used to analyze cellular gene expression and to facilitate tracking of cell lineages in vivo. One of these, enhanced yellow fluorescent protein (EYFP) has several properties such as intense fluorescence and little to no toxicity in cells, which makes it an excellent molecule to label proteins and cells of interest. In live cells, visualization of EYFP has been highly successful; however, detection of EYFP in lymphoid tissue sections, particularly in combination with other markers of interest has been difficult. This is because of the enhanced solubility of EYFP in the absence of fixation. When extended fixation protocols are employed, EYFP is preserved but detection of other cellular antigens becomes problematic due to over fixation. Here we demonstrate that EYFP-expressing T and B cells can be efficiently visualized in lymphoid tissue sections without compromising the ability to detect other cellular markers.
Collapse
Affiliation(s)
- Joseph G Dauner
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Center, Emory University, Atlanta, GA 30329, United States
| | | | | | | |
Collapse
|
562
|
Zhu B, Symonds ALJ, Martin JE, Kioussis D, Wraith DC, Li S, Wang P. Early growth response gene 2 (Egr-2) controls the self-tolerance of T cells and prevents the development of lupuslike autoimmune disease. J Exp Med 2008; 205:2295-307. [PMID: 18779345 PMCID: PMC2556781 DOI: 10.1084/jem.20080187] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 08/07/2008] [Indexed: 11/08/2022] Open
Abstract
Maintaining tolerance of T cells to self-antigens is essential to avoid autoimmune disease. How self-reactive T cells are kept functionally inactive is, however, unknown. In this study, we show that early growth response gene 2 (Egr-2), a zinc-finger transcription factor, is expressed in CD44(high) T cells and controls their proliferation and activation. In the absence of Egr-2, CD44(high), but not CD44(low) T cells, are hyperreactive and hyperproliferative in vivo. The accumulation of activated CD4(+)CD44(high) T cells leads to the development of a late onset lupuslike autoimmune disease characterized by the accumulation of interferon (IFN)-gamma and interleukin (IL)-17-producing CD4(+) T cells, loss of tolerance to nuclear antigens, massive infiltration of T cells into multiple organs and glomerulonephritis. We found that the expression of cyclin-dependent kinase inhibitor p21cip1 was impaired in Egr-2-deficient T cells, whereas the expression of IFN-gamma and IL-17 in response to T cell receptor ligation was significantly increased, suggesting that Egr-2 activates the expression of genes involved in the negative regulation of T cell proliferation and inflammation. These results demonstrate that Egr-2 is an intrinsic regulator of effector T cells and controls the expansion of self-reactive T cells and development of autoimmune disease.
Collapse
Affiliation(s)
- Bo Zhu
- Institute of Cell and Molecular Science, Barts and London School of Medicine and Dentistry, University of London, London E1 2AT, England, UK
| | | | | | | | | | | | | |
Collapse
|
563
|
Walkley CR, Sankaran VG, Orkin SH. Rb and hematopoiesis: stem cells to anemia. Cell Div 2008; 3:13. [PMID: 18775080 PMCID: PMC2562376 DOI: 10.1186/1747-1028-3-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Accepted: 09/08/2008] [Indexed: 12/31/2022] Open
Abstract
The retinoblastoma protein, Rb, was one of the first tumor suppressor genes identified as a result of the familial syndrome retinoblastoma. In the period since its identification and cloning a large number of studies have described its role in various cellular processes. The application of conditional somatic mutation with lineage and temporally controlled gene deletion strategies, thus circumventing the lethality associated with germ-line deletion of Rb, have allowed for a reanalysis of the in vivo role of Rb. In the hematopoietic system, such approaches have led to new insights into stem cell biology and the role of the microenvironment in regulating hematopoietic stem cell fate. They have also clarified the role that Rb plays during erythropoiesis and defined a novel mechanism linking mitochondrial function to terminal cell cycle withdrawal. These studies have shed light on the in vivo role of Rb in the regulation of hematopoiesis and also prompt further analysis of the role that Rb plays in both the regulation of hematopoietic stem cells and the terminal differentiation of their progeny.
Collapse
Affiliation(s)
- Carl R Walkley
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology and Stem Cell Program, Children's Hospital Boston, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA.,St. Vincent's Institute, Department of Medicine at St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Vijay G Sankaran
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology and Stem Cell Program, Children's Hospital Boston, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Stuart H Orkin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology and Stem Cell Program, Children's Hospital Boston, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA.,Howard Hughes Medical Institute, Boston, MA, 02115, USA
| |
Collapse
|
564
|
Ananieva O, Macdonald A, Wang X, McCoy CE, McIlrath J, Tournier C, Arthur JSC. ERK5 regulation in naïve T-cell activation and survival. Eur J Immunol 2008; 38:2534-47. [PMID: 18792406 DOI: 10.1002/eji.200737867] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
ERK5 has been implicated in regulating the MEF2-dependent genes Klf2 and nur77 downstream of the TCR and the maintenance of expression of CD62L on peripheral T cells. Based on this data, knockout of ERK5 would be predicted to compromise T-cell development and the maintenance of T cells in the periphery. Using an ERK5 conditional knockout, driven by CD4-CRE or Vav-CRE transgenes resulting in the loss of ERK5 in T cells, we have found that ERK5 is not required for T-cell development. In addition, normal numbers of T cells were found in the spleens and lymph nodes of these mice. We also find that TCR stimulation is not a strong signal for ERK5 activation in primary murine T cells. ERK5 was found to contribute to the induction of Klf2 but not nur77 mRNA following TCR activation. Despite the reduction in Klf2 mRNA, no effect was seen in ERK5 knockouts on either the mRNA levels for the Klf2 target genes CD62L, CCR7 and S1P, or the cell surface expression of CD62L. These results suggest that while ERK5 does contribute to Klf2 regulation in T cells, it is not essential for the expression of CD62L or T-cell survival.
Collapse
Affiliation(s)
- Olga Ananieva
- MRC Protein Phosphorylation Unit, College of Life Sciences, Sir James Black Centre, University of Dundee, Dundee, UK
| | | | | | | | | | | | | |
Collapse
|
565
|
Hou B, Reizis B, DeFranco AL. Toll-like receptors activate innate and adaptive immunity by using dendritic cell-intrinsic and -extrinsic mechanisms. Immunity 2008; 29:272-82. [PMID: 18656388 PMCID: PMC2847796 DOI: 10.1016/j.immuni.2008.05.016] [Citation(s) in RCA: 305] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 05/13/2008] [Accepted: 05/23/2008] [Indexed: 01/24/2023]
Abstract
Toll-like receptors (TLRs) play prominent roles in initiating immune responses to infection, but their roles in particular cell types in vivo are not established. Here we report the generation of mice selectively lacking the crucial TLR-signaling adaptor MyD88 in dendritic cells (DCs). In these mice, the early production of inflammatory cytokines, especially IL-12, was substantially reduced after TLR stimulation. Whereas the innate interferon-gamma response of natural killer cells and of natural killer T cells and the Th1 polarization of antigen-specific CD4(+) T cells were severely compromised after treatment with a soluble TLR9 ligand, they were largely intact after administration of an aggregated TLR9 ligand. These results demonstrate that the physical form of a TLR ligand affects which cells can respond to it and that DCs and other innate immune cells can respond via TLRs and collaborate in promoting Th1 adaptive immune responses to an aggregated stimulus.
Collapse
Affiliation(s)
- Baidong Hou
- Department of Microbiology & Immunology, University of California, San Francisco, CA, 94143, USA
| | | | | |
Collapse
|
566
|
Maroof A, Beattie L, Zubairi S, Svensson M, Stager S, Kaye PM. Posttranscriptional regulation of II10 gene expression allows natural killer cells to express immunoregulatory function. Immunity 2008; 29:295-305. [PMID: 18701085 PMCID: PMC2656759 DOI: 10.1016/j.immuni.2008.06.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 04/21/2008] [Accepted: 06/06/2008] [Indexed: 01/13/2023]
Abstract
Natural killer (NK) cells play a well-recognized role in early pathogen containment and in shaping acquired cell-mediated immunity. However, indirect evidence in humans and experimental models has suggested that NK cells also play negative regulatory roles during chronic disease. To formally test this hypothesis, we employed a well-defined experimental model of visceral leishmaniasis. Our data demonstrated that NKp46(+)CD49b(+)CD3(-) NK cells were recruited to the spleen and into hepatic granulomas, where they inhibited host protective immunity in an interleukin-10 (IL-10)-dependent manner. Although IL-10 mRNA could be detected in activated NK cells 24 hr after infection, the inhibitory function of NK cells was only acquired later during infection, coincident with increased IL-10 mRNA stability and an enhanced capacity to secrete IL-10 protein. Our data support a growing body of literature that implicates NK cells as negative regulators of cell-mediated immunity and suggest that NK cells, like CD4(+) T helper 1 cells, may acquire immunoregulatory functions as a consequence of extensive activation.
Collapse
Affiliation(s)
- Asher Maroof
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| | - Lynette Beattie
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| | - Soombul Zubairi
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| | - Mattias Svensson
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| | - Simona Stager
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| | - Paul M. Kaye
- Immunology and Infection Unit, Hull York Medical School and Dept. of Biology, University of York, Wentworth Way, York YO10 5YW UK
| |
Collapse
|
567
|
Akins EJ, Dubey P. Noninvasive imaging of cell-mediated therapy for treatment of cancer. J Nucl Med 2008; 49 Suppl 2:180S-95S. [PMID: 18523073 DOI: 10.2967/jnumed.107.045971] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-mediated therapy (immunotherapy) for the treatment of cancer is an active area of investigation in animal models and clinical trials. Despite many advances, objective responses to immunotherapy are observed in a small number of cases, for certain tumor types. To better understand differences in outcomes, it is critical to develop assays for tracking effector cell localization and function in situ. The fairly recent use of molecular imaging techniques to track cell populations has presented researchers and clinicians with a powerful diagnostic tool for determining the efficacy of cell-mediated therapy for the treatment of cancer. This review highlights the application of whole-body noninvasive radioisotopic, magnetic, and optical imaging methods for monitoring effector cells in vivo. Issues that affect sensitivity of detection, such as methods of cell marking, efficiency of cell labeling, toxicity, and limits of detection of imaging modalities, are discussed.
Collapse
Affiliation(s)
- Elizabeth J Akins
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
568
|
He C, Hu H, Braren R, Fong SY, Trumpp A, Carlson TR, Wang RA. c-myc in the hematopoietic lineage is crucial for its angiogenic function in the mouse embryo. Development 2008; 135:2467-77. [PMID: 18550710 PMCID: PMC2597486 DOI: 10.1242/dev.020131] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The c-myc proto-oncogene, which is crucial for the progression of many human cancers, has been implicated in key cellular processes in diverse cell types, including endothelial cells that line the blood vessels and are critical for angiogenesis. The de novo differentiation of endothelial cells is known as vasculogenesis, whereas the growth of new blood vessels from pre-existing vessels is known as angiogenesis. To ascertain the function of c-myc in vascular development, we deleted c-myc in selected cell lineages. Embryos lacking c-myc in endothelial and hematopoietic lineages phenocopied those lacking c-myc in the entire embryo proper. At embryonic day (E) 10.5, both mutant embryos were grossly normal, had initiated primitive hematopoiesis, and both survived until E11.5-12.5, longer than the complete null. However, they progressively developed defective hematopoiesis and angiogenesis. The majority of embryos lacking c-myc specifically in hematopoietic cells phenocopied those lacking c-myc in endothelial and hematopoietic lineages, with impaired definitive hematopoiesis as well as angiogenic remodeling. c-myc is required for embryonic hematopoietic stem cell differentiation, through a cell-autonomous mechanism. Surprisingly, c-myc is not required for vasculogenesis in the embryo. c-myc deletion in endothelial cells does not abrogate endothelial proliferation, survival, migration or capillary formation. Embryos lacking c-myc in a majority of endothelial cells can survive beyond E12.5. Our findings reveal that hematopoiesis is a major function of c-myc in embryos and support the notion that c-myc functions in selected cell lineages rather than in a ubiquitous manner in mammalian development.
Collapse
Affiliation(s)
- Chen He
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| | - Huiqing Hu
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| | - Rickmer Braren
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| | - Shun-Yin Fong
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| | - Andreas Trumpp
- Genetics and Stem Cell Laboratory, Swiss Institute for Experimental Cancer Research, Ch. des Boveresses 155, CH-1066, Epalinges, Switzerland Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, CH-1015 Lausanne, Switzerland
| | - Timothy R. Carlson
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| | - Rong A. Wang
- Pacific Vascular Research Laboratory, Division of Vascular Surgery, Departments of Surgery and Anatomy, University of California, San Francisco, CA 94143
| |
Collapse
|
569
|
Kwon K, Hutter C, Sun Q, Bilic I, Cobaleda C, Malin S, Busslinger M. Instructive role of the transcription factor E2A in early B lymphopoiesis and germinal center B cell development. Immunity 2008; 28:751-62. [PMID: 18538592 DOI: 10.1016/j.immuni.2008.04.014] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2007] [Revised: 03/12/2008] [Accepted: 04/01/2008] [Indexed: 02/06/2023]
Abstract
The transcription factor E2A controls the initiation of B lymphopoiesis, which is arrested at the pre-pro-B cell stage in E2A-deficient mice. Here, we demonstrate by conditional mutagenesis that E2A is essential for the development of pro-B, pre-B, and immature B cells in the bone marrow. E2A is, however, dispensable for the generation of mature B cells and plasma cells in peripheral lymphoid organs. In contrast, germinal center B cell development is impaired in the absence of E2A despite normal AID expression and class-switch recombination. Molecular analysis revealed that E2A is required not only for initiating but also for maintaining the expression of Ebf1, Pax5, and the B cell gene program in pro-B cells. Notably, precocious Pax5 transcription from the Ikzf1 locus promotes pro-B cell development in E2A-deficient mice, demonstrating that ectopic Pax5 expression is sufficient to activate the B lymphoid transcription program in vivo in the absence of E2A.
Collapse
Affiliation(s)
- Kyongrim Kwon
- Research Institute of Molecular Pathology, Vienna Biocenter, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
570
|
Martin A, Aguirre J, Sarasa-Renedo A, Tsoukatou D, Garofalakis A, Meyer H, Mamalaki C, Ripoll J, Planas AM. Imaging Changes in Lymphoid Organs In Vivo after Brain Ischemia with Three-Dimensional Fluorescence Molecular Tomography in Transgenic Mice Expressing Green Fluorescent Protein in T Lymphocytes. Mol Imaging 2008. [DOI: 10.2310/7290.2008.00016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Abraham Martin
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Juan Aguirre
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Ana Sarasa-Renedo
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Debbie Tsoukatou
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Anikitos Garofalakis
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Heiko Meyer
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Clio Mamalaki
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Jorge Ripoll
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Anna M. Planas
- From the Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain, and Foundation for Research and Technology-Hellas, Institute of Electronic Structure and Laser and Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| |
Collapse
|
571
|
Shi M, Lin TH, Appell KC, Berg LJ. Janus-kinase-3-dependent signals induce chromatin remodeling at the Ifng locus during T helper 1 cell differentiation. Immunity 2008; 28:763-73. [PMID: 18549798 PMCID: PMC2587400 DOI: 10.1016/j.immuni.2008.04.016] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 01/06/2023]
Abstract
Differentiation of naive CD4+ T cells into T helper type 1 (Th1) effector cells requires both T cell receptor (TCR) signaling and cytokines such as interleukin-12 and interferon gamma (IFN-gamma). Here, we report that a third cytokine signal, mediated by the Janus family tyrosine kinase 3 (Jak3) and signal transducer and activator of transcription 5 (STAT5) pathway, is also required for Th1 cell differentiation. In the absence of Jak3-dependent signals, naive CD4+ T cells proliferate robustly but produce little IFN-gamma after Th1 cell polarization in vitro. This defect is not due to reduced activation of STAT1 or STAT4 or to impaired upregulation of the transcription factor T-bet. Instead, we find that T-bet binding to the Ifng promoter is greatly diminished in the absence of Jak3-dependent signals, correlating with a decrease in Ifng promoter accessibility and histone acetylation. These data indicate that Jak3 regulates epigenetic modification and chromatin remodeling of the Ifng locus during Th1 cell differentiation.
Collapse
Affiliation(s)
- Min Shi
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
572
|
Chen D, Abrahams JM, Smith LM, McVey JH, Lechler RI, Dorling A. Regenerative repair after endoluminal injury in mice with specific antagonism of protease activated receptors on CD34+ vascular progenitors. Blood 2008; 111:4155-64. [PMID: 18268094 DOI: 10.1182/blood-2007-10-120295] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tissue factor (TF) and thrombin are involved in intimal hyperplasia (IH) and remodelling following vascular injury. Because many neointimal smooth muscle cells (VSMCs) derive from circulating vascular progenitors (VPs), we investigated how thrombin influences VP phenotype and function. Following wire-induced carotid artery injury in mice, the majority of circulating VPs expressed TF, were capable of initiating clotting in vitro, and had protease-activated receptors (PAR)-1, -2, and -4. Thrombin, through PAR-1, inhibited apoptosis and caused proliferation, resulting in the outgrowth of VP coexpressing markers of activated endothelial cells and VSMCs, even in the presence of growth factors. These mixed-phenotype VPs circulated as a minority population after injury and shared a similar phenotype with many neointimal cells. Labeled CD34(+) cells, injected up to 2 weeks after injury, could be detected in the injured vessel wall, suggesting that continued recruitment may contribute to progressive IH. Finally, CD34(+) cells incubated with thrombin prior to injection promoted florid neointimal lesions, whereas those incubated with PAR antagonists inhibited IH and promoted regenerative repair characterized by the development of a quiescent endothelium. We conclude that IH after vascular injury is due to the direct actions of thrombin on mobilized VPs.
Collapse
Affiliation(s)
- Daxin Chen
- Department of Immunology, Imperial College London, Hammersmith Hospital, London, UK
| | | | | | | | | | | |
Collapse
|
573
|
Xiao C, Srinivasan L, Calado DP, Patterson HC, Zhang B, Wang J, Henderson JM, Kutok JL, Rajewsky K. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat Immunol 2008; 9:405-14. [PMID: 18327259 PMCID: PMC2533767 DOI: 10.1038/ni1575] [Citation(s) in RCA: 991] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Accepted: 02/13/2008] [Indexed: 02/07/2023]
Abstract
The genomic region encoding the miR-17-92 microRNA (miRNA) cluster is often amplified in lymphoma and other cancers, and cancer cells carrying this amplification have higher expression of miRNA in this cluster. Retroviral expression of miR-17-92 accelerates c-Myc-induced lymphoma development, but precisely how higher expression of miR-17-92 promotes lymphomagenesis remains unclear. Here we generated mice with higher expression of miR-17-92 in lymphocytes. These mice developed lymphoproliferative disease and autoimmunity and died prematurely. Lymphocytes from these mice showed more proliferation and less activation-induced cell death. The miR-17-92 miRNA suppressed expression of the tumor suppressor PTEN and the proapoptotic protein Bim. This mechanism probably contributed to the lymphoproliferative disease and autoimmunity of miR-17-92-transgenic mice and contributes to lymphoma development in patients with amplifications of the miR-17-92 coding region.
Collapse
Affiliation(s)
- Changchun Xiao
- Immune Disease Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
574
|
Zha Y, Shah R, Locke F, Wong A, Gajewski TF. Use of Cre-adenovirus and CAR transgenic mice for efficient deletion of genes in post-thymic T cells. J Immunol Methods 2008; 331:94-102. [PMID: 18177887 PMCID: PMC3278801 DOI: 10.1016/j.jim.2007.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 10/22/2007] [Accepted: 11/25/2007] [Indexed: 11/15/2022]
Abstract
Conditional gene deletion using lineage-specific transgenic expression of Cre has been useful for defining the role of specific gene products in mice in vivo. However, this technology has had limitations for studies of peripheral T cell biology, since the T-lineage promoters commonly used are active early in thymic development. As such, T cell development can be altered by the resulting genetic alterations, thus limiting the interpretation of the data in post-thymic T cell studies. Thus, new strategies are needed to delete targeted genes directly in peripheral T lymphocytes. The availability of transgenic mice expressing the CAR in the T cell compartment enabled testing of Cre-mediated recombination using an adenoviral vector in naïve peripheral T cells in vitro, even without cellular activation. Using Rosa26R reporterxCAR transgenic mice, we describe conditions by which Cre-mediated deletion of targeted genes can be achieved with primary T cells in vitro. These cells can also be adoptively transferred into defined recipient mice for study in vivo. We use conditional PTEN-deficient mice as proof of concept to confirm the value of this technique for deleting a negative regulator of T cell activation. This technology should be broadly applicable for studies of T cell-specific gene deletion to gain understanding of function in the post-thymic T cell compartment.
Collapse
Affiliation(s)
- Yuanyuan Zha
- Department of Pathology and Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Ramila Shah
- Department of Pathology and Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Frederick Locke
- Department of Pathology and Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Austin Wong
- Department of Pathology and Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Thomas F. Gajewski
- Department of Pathology and Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
575
|
Dumont C, Henderson R, Tybulewicz VLJ. Characterization of the roles of Rac1 and Rac2 GTPases in lymphocyte development. Methods Enzymol 2008; 439:235-54. [PMID: 18374169 DOI: 10.1016/s0076-6879(07)00418-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This chapter describes methods for the analysis of B and T lymphocyte development in mice deficient in Rac1 and/or Rac2 GTPases. The development of both B and T cells is critically dependent on transition through checkpoints monitoring for correct rearrangement of antigen receptor genes. Progression through these checkpoints depends on signaling from the antigen receptors. In addition, signals from cytokine, chemokine, Notch, and death receptors play important roles in the survival, proliferation, and migration of developing lymphocytes. Analysis of these processes in mice deficient in these GTPases can illuminate their roles in transducing signals from these different receptors.
Collapse
Affiliation(s)
- Celine Dumont
- Division of Immune Cell Biology, National Institute for Medical Research, London, United Kingdom
| | | | | |
Collapse
|
576
|
Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood 2007; 111:3931-40. [PMID: 18160670 DOI: 10.1182/blood-2007-08-107748] [Citation(s) in RCA: 344] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
An acquired somatic mutation in the JAK2 gene (JAK2-V617F) is present in the majority of patients with myeloproliferative disorders (MPDs). Several phenotypic manifestations (polycythemia vera [PV], essential thrombocythemia [ET], and primary myelofibrosis) can be associated with the same mutation. We generated JAK2-V617F transgenic mice using a human JAK2 gene with the sequences encoding the kinase domain placed in the inverse orientation and flanked by antiparallel loxP sites. Crossing mice of one transgenic line (FF1) with transgenic mice expressing Cre-recombinase under the control of the hematopoiesis specific Vav promoter led to expression of JAK2-V617F that was lower than the endogenous wild-type Jak2. These mice developed a phenotype resembling ET with strongly elevated platelet counts and moderate neutrophilia. Induction of the JAK2-V617F transgene with the interferon-inducible MxCre resulted in expression of JAK2-V617F approximately equal to wild-type Jak2 and a PV-like phenotype with increased hemoglobin, thrombocytosis, and neutrophilia. Higher levels of JAK2-V617F in mouse bone marrow by retroviral transduction caused a PV-like phenotype without thrombocytosis. These data are consistent with the hypothesis that the ratio of mutant to wild-type JAK2 is critical for the phenotypic manifestation. A similar correlation was also found in patients with MPD.
Collapse
|
577
|
Zhao C, Blum J, Chen A, Kwon HY, Jung SH, Cook JM, Lagoo A, Reya T. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell 2007; 12:528-41. [PMID: 18068630 PMCID: PMC2262869 DOI: 10.1016/j.ccr.2007.11.003] [Citation(s) in RCA: 471] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 06/15/2007] [Accepted: 11/02/2007] [Indexed: 12/16/2022]
Abstract
A key characteristic of stem cells and cancer cells is their ability to self-renew. To test if Wnt signaling can regulate the self-renewal of both stem cells and cancer cells in the hematopoietic system, we developed mice that lack beta-catenin in their hematopoietic cells. Here we show that beta-catenin-deficient mice can form HSCs, but that these cells are deficient in long-term growth and maintenance. Moreover, beta-catenin deletion causes a profound reduction in the ability of mice to develop BCR-ABL-induced chronic myelogenous leukemia (CML), while allowing progression of acute lymphocytic leukemia (ALL). These studies demonstrate that Wnt signaling is required for the self-renewal of normal and neoplastic stem cells in the hematopoietic system.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jordan Blum
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alan Chen
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hyog Young Kwon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Seung Hye Jung
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - J. Michael Cook
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Anand Lagoo
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Tannishtha Reya
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
578
|
Hooper J, Maurice D, Argent-Katwala MJG, Weston K. Myb proteins regulate expression of histone variant H2A.Z during thymocyte development. Immunology 2007; 123:282-9. [PMID: 17931383 DOI: 10.1111/j.1365-2567.2007.02697.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The c-myb gene encodes a transcription factor required for the normal development of T cells in the thymus, and for subsequent peripheral T-cell activation and survival. However, the profile of genes known to be transcriptionally regulated by c-Myb in T cells does not adequately explain the pleiotrophic nature of the effects of c-Myb. We present here a detailed molecular characterization of the regulation of a novel target gene, the histone variant H2A.Z. We show that c-Myb is able to bind to and activate the H2A.Z promoter in T cells both in vitro and in vivo, and present evidence that perturbation of Myb activity during T-cell development results in reduced H2A.Z expression. As H2A.Z is absolutely required for the early stages of mammalian development, and plays essential roles in the regulation of chromatin structure in gene promoters in yeast, its regulation by c-Myb is likely to be of some importance during T-cell development.
Collapse
Affiliation(s)
- Joel Hooper
- Institute of Cancer Research, CRUK Centre for Cell and Molecular Biology, London, UK
| | | | | | | |
Collapse
|
579
|
Travis MA, Reizis B, Melton AC, Masteller E, Tang Q, Proctor JM, Wang Y, Bernstein X, Huang X, Reichardt LF, Bluestone JA, Sheppard D. Loss of integrin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice. Nature 2007; 449:361-5. [PMID: 17694047 PMCID: PMC2670239 DOI: 10.1038/nature06110] [Citation(s) in RCA: 419] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Accepted: 07/23/2007] [Indexed: 02/08/2023]
Abstract
The cytokine transforming growth factor-beta (TGF-beta) is an important negative regulator of adaptive immunity. TGF-beta is secreted by cells as an inactive precursor that must be activated to exert biological effects, but the mechanisms that regulate TGF-beta activation and function in the immune system are poorly understood. Here we show that conditional loss of the TGF-beta-activating integrin alpha(v)beta8 on leukocytes causes severe inflammatory bowel disease and age-related autoimmunity in mice. This autoimmune phenotype is largely due to lack of alpha(v)beta8 on dendritic cells, as mice lacking alpha(v)beta8 principally on dendritic cells develop identical immunological abnormalities as mice lacking alpha(v)beta8 on all leukocytes, whereas mice lacking alpha(v)beta8 on T cells alone are phenotypically normal. We further show that dendritic cells lacking alpha(v)beta8 fail to induce regulatory T cells (T(R) cells) in vitro, an effect that depends on TGF-beta activity. Furthermore, mice lacking alpha(v)beta8 on dendritic cells have reduced proportions of T(R) cells in colonic tissue. These results suggest that alpha(v)beta8-mediated TGF-beta activation by dendritic cells is essential for preventing immune dysfunction that results in inflammatory bowel disease and autoimmunity, effects that are due, at least in part, to the ability of alpha(v)beta8 on dendritic cells to induce and/or maintain tissue T(R) cells.
Collapse
Affiliation(s)
- Mark A Travis
- Lung Biology Center, Department of Medicine, University of California San Francisco, 1550 4th Street, Room 545, San Francisco, California 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
580
|
McMahon KA, Hiew SYL, Hadjur S, Veiga-Fernandes H, Menzel U, Price AJ, Kioussis D, Williams O, Brady HJM. Mll has a critical role in fetal and adult hematopoietic stem cell self-renewal. Cell Stem Cell 2007; 1:338-45. [PMID: 18371367 DOI: 10.1016/j.stem.2007.07.002] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 05/22/2007] [Accepted: 07/06/2007] [Indexed: 11/15/2022]
Abstract
The Mixed Lineage Leukemia (Mll) gene is a homolog of Drosophila Trithorax commonly rearranged in infant leukemia. Comprehensive analysis of the role of Mll in hematopoiesis in fetal and adult knockout mice has been prevented by the lethality of Mll(-/-) mice. We have established a conditional deletion model that allows us to study adult hematopoiesis in the absence of Mll. In this study, Mll(-/-) embryos survive to E16.5 and have reduced numbers of HSCs. The quiescent fraction of these HSCs is greatly reduced, and they are unable to compete with wild-type cells in transplantation assays. Mice with Mll expression conditionally deleted in the hematopoietic system have grossly normal hematopoiesis in bone marrow, thymus, and spleen. However, transplanted Mll-deficient bone marrow cells are highly compromised in their ability to competitively reconstitute irradiated recipients. These results suggest a critical role for Mll in regulating stem cell self-renewal.
Collapse
Affiliation(s)
- Kathryn A McMahon
- Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, WC1N 1EH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
581
|
Pan D, Schomber T, Kalberer CP, Terracciano LM, Hafen K, Krenger W, Hao-Shen H, Deng C, Skoda RC. Normal erythropoiesis but severe polyposis and bleeding anemia in Smad4-deficient mice. Blood 2007; 110:3049-55. [PMID: 17638848 DOI: 10.1182/blood-2007-02-074393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The tumor suppressor Smad4 mediates signaling by the transforming growth factor beta (TGF-beta) superfamily of ligands. Previous studies showed that several TGF-beta family members exert important functions in hematopoiesis. Here, we studied the role of Smad4 in adult murine hematopoiesis using the inducible Mx-Cre/loxP system. Mice with homozygous Smad4 deletion (Smad4(Delta/Delta)) developed severe anemia 6 to 8 weeks after induction (mean hemoglobin level 70 g/L). The anemia was not transplantable, as wild-type mice reconstituted with Smad4(Delta/Delta) bone marrow cells had normal peripheral blood counts. These mice did not develop an inflammatory disease typical for mice deficient in TGF-beta receptors I and II, suggesting that the suppression of inflammation by TGF-beta is Smad4 independent. The same results were obtained when Smad4 alleles were deleted selectively in hematopoietic cells using the VavCre transgenic mice. In contrast, lethally irradiated Smad4(Delta/Delta) mice that received wild-type bone marrow cells developed anemia similar to Smad4(Delta/Delta) mice that did not receive a transplant. Liver iron stores were decreased and blood was present in stool, indicating that the anemia was due to blood loss. Multiple polyps in stomach and colon represent a likely source of the bleeding. We conclude that Smad4 is not required for adult erythropoiesis and that anemia is solely the consequence of blood loss.
Collapse
Affiliation(s)
- Dejing Pan
- Department of Research, Experimental Hematology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
582
|
Meyer H, Garofalakis A, Zacharakis G, Psycharakis S, Mamalaki C, Kioussis D, Economou EN, Ntziachristos V, Ripoll J. Noncontact optical imaging in mice with full angular coverage and automatic surface extraction. APPLIED OPTICS 2007; 46:3617-27. [PMID: 17514324 DOI: 10.1364/ao.46.003617] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
During the past decade, optical imaging combined with tomographic approaches has proved its potential in offering quantitative three-dimensional spatial maps of chromophore or fluorophore concentration in vivo. Due to its direct application in biology and biomedicine, diffuse optical tomography (DOT) and its fluorescence counterpart, fluorescence molecular tomography (FMT), have benefited from an increase in devoted research and new experimental and theoretical developments, giving rise to a new imaging modality. The most recent advances in FMT and DOT are based on the capability of collecting large data sets by using CCDs as detectors, and on the ability to include multiple projections through recently developed noncontact approaches. For these to be implemented, we have developed an imaging setup that enables three-dimensional imaging of arbitrary shapes in fluorescence or absorption mode that is appropriate for small animal imaging. This is achieved by implementing a noncontact approach both for sources and detectors and coregistering surface geometry measurements using the same CCD camera. A thresholded shadowgrammetry approach is applied to the geometry measurements to retrieve the surface mesh. We present the evaluation of the system and method in recovering three-dimensional surfaces from phantom data and live mice. The approach is used to map the measured in vivo fluorescence data onto the tissue surface by making use of the free-space propagation equations, as well as to reconstruct fluorescence concentrations inside highly scattering tissuelike phantom samples. Finally, the potential use of this setup for in vivo small animal imaging and its impact on biomedical research is discussed.
Collapse
Affiliation(s)
- Heiko Meyer
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, Heraklion Crete, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
583
|
Kim WI, Wiesner SM, Largaespada DA. Vav promoter-tTA conditional transgene expression system for hematopoietic cells drives high level expression in developing B and T cells. Exp Hematol 2007; 35:1231-9. [PMID: 17560009 DOI: 10.1016/j.exphem.2007.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 04/10/2007] [Accepted: 04/20/2007] [Indexed: 11/24/2022]
Abstract
OBJECTIVE We previously showed that Vav promoter-tetracycline transactivator (Vav-tTA)-driven tetracycline-regulated element (TRE)-NRAS(V12) expression resulted in mastocytosis development in mice. To investigate which hematopoietic cells express TRE-driven transgenes when combined with Vav-tTA, we assayed hematopoietic cells, including bone marrow-derived mast cells (BMMC) and CD34-positive hematopoietic progenitor cells (HPC) as well as myeloid and lymphoid lineages. To determine if suppression of NRAS(V12) expression early in life would delay mastocytosis we treated developing and juvenile mice with doxycycline (Dox). MATERIALS AND METHODS Vav-tTA-driven luciferase expression was assayed by live mouse imaging and relative light unit measurement before or after treating Vav-tTA and TRE-luciferase (TRE-Luc) cotransgenic mice with Dox. Magnetic cell sorting and fluorescence-activating cell sorting methods were used to sort hematopoietic cells. To suppress TRE-mediated luciferase or NRAS(V12) expression in Vav-tTA cotransgenic mice, we added Dox to the drinking water. RESULTS B cells in the bone marrow and T cells in the thymus expressed Vav-tTA-driven luciferase at much higher levels than in myeloid cells, BMMC, and CD34-positive HPC, which showed relatively low levels. Dox treatment completely eliminated the luciferase expression from all hematopoietic cells. Repression of TRE-NRAS(V12) expression early in life was sufficient to increase the latency of mastocytosis development. CONCLUSION The Vav-tTA transgenic line will be very useful for conditional transgene expression in developing B and T cells. Vav-tTA-driven NRAS(V12) expression is sufficient for mastocytosis development, but not for myeloid leukemia. Lymphoid cells are resistant to NRAS(V12) transformation despite high level of expression.
Collapse
Affiliation(s)
- Won-Il Kim
- Graduate Program in Molecular, Cellular, Developmental Biology and Genetics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
584
|
Chatzidakis I, Fousteri G, Tsoukatou D, Kollias G, Mamalaki C. An Essential Role for TNF in Modulating Thresholds for Survival, Activation, and Tolerance of CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:6735-45. [PMID: 17513720 DOI: 10.4049/jimmunol.178.11.6735] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
TNF and its receptors p55 and p75 are known to be important in the homeostasis of the peripheral immune system. Previous studies have presented apparently contradictory evidence for an in vivo role of TNF in T cells. In this study, we analyzed TNF-deficient mice crossed with the F5 TCR-transgenic animals. We show that endogenous TNF modulates several aspects of homeostasis of peripheral F5 CD8 T cells. We found that F5/TNF(-/-)mice had reduced numbers of peripheral F5 T cells, F5/TNF(-/-) CD8 T cells exhibited reduced survival potential, and furthermore that T cell-derived TNF is required for optimum recovery of naive CD8 T cells in lymphopenic hosts, suggesting its involvement in the survival of peripheral CD8 T cells. Both peptide activation and ensuing Ag-induced apoptosis are quantitatively reduced in TNF(-/-) CD8 T cells. The latter observations can be related to decreased binding activities of NF-kappaB and NF-ATp observed in Ag-stimulated F5/TNF(-/-) T cells. Finally, in a CD8 T cell tolerance model, endogenous TNF was necessary for several parameters of CD8 T cell tolerance induction. Collectively, our results provide evidence that endogenous TNF modulates thresholds in several ligand-driven T cell responses.
Collapse
Affiliation(s)
- Ioannis Chatzidakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Vassilika Vouton, Crete, Greece
| | | | | | | | | |
Collapse
|
585
|
Veiga-Fernandes H, Coles MC, Foster KE, Patel A, Williams A, Natarajan D, Barlow A, Pachnis V, Kioussis D. Tyrosine kinase receptor RET is a key regulator of Peyer's patch organogenesis. Nature 2007; 446:547-51. [PMID: 17322904 DOI: 10.1038/nature05597] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Accepted: 01/15/2007] [Indexed: 11/09/2022]
Abstract
Normal organogenesis requires co-ordinate development and interaction of multiple cell types, and is seemingly governed by tissue specific factors. Lymphoid organogenesis during embryonic life is dependent on molecules the temporal expression of which is tightly regulated. During this process, haematopoietic 'inducer' cells interact with stromal 'organizer' cells, giving rise to the lymphoid organ primordia. Here we show that the haematopoietic cells in the gut exhibit a random pattern of motility before aggregation into the primordia of Peyer's patches, a major component of the gut-associated lymphoid tissue. We further show that a CD45+CD4-CD3-Il7Ralpha-c-Kit+CD11c+ haematopoietic population expressing lymphotoxin has an important role in the formation of Peyer's patches. A subset of these cells expresses the receptor tyrosine kinase RET, which is essential for mammalian enteric nervous system formation. We demonstrate that RET signalling is also crucial for Peyer's patch formation. Functional genetic analysis revealed that Gfra3-deficiency results in impairment of Peyer's patch development, suggesting that the signalling axis RET/GFRalpha3/ARTN is involved in this process. To support this hypothesis, we show that the RET ligand ARTN is a strong attractant of gut haematopoietic cells, inducing the formation of ectopic Peyer's patch-like structures. Our work strongly suggests that the RET signalling pathway, by regulating the development of both the nervous and lymphoid system in the gut, has a key role in the molecular mechanisms that orchestrate intestine organogenesis.
Collapse
Affiliation(s)
- Henrique Veiga-Fernandes
- Division of Molecular Immunology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
586
|
Garofalakis A, Zacharakis G, Meyer H, Economou EN, Mamalaki C, Papamatheakis J, Kioussis D, Ntziachristos V, Ripoll J. Three-Dimensional in Vivo Imaging of Green Fluorescent Protein-Expressing T Cells in Mice with Noncontact Fluorescence Molecular Tomography. Mol Imaging 2007. [DOI: 10.2310/7290.2007.00007] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Anikitos Garofalakis
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Giannis Zacharakis
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Heiko Meyer
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Eleftherios N. Economou
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Clio Mamalaki
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Joseph Papamatheakis
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Dimitris Kioussis
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Vasilis Ntziachristos
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| | - Jorge Ripoll
- From the Institutes of Electronic Structure and Laser and Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion Crete, Greece; Division of Molecular Immunology, National Institute for Medical Research, London, UK; and Laboratory for Bio-Optics and Molecular Imaging, Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA
| |
Collapse
|
587
|
Roes J. Conditional mutagenesis reveals immunological functions of widely expressed genes: activation thresholds, homeostatic mechanisms and disease models. Handb Exp Pharmacol 2007:289-314. [PMID: 17203660 DOI: 10.1007/978-3-540-35109-2_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Evolutionarily conserved, widely expressed genes provide the functional backbone of most, if not all, cell types. Although mouse mutants created by germ line gene inactivation are instrumental in establishing the importance of such genes in vivo, distortion of embryonic development or multiple body systems often preclude detailed functional studies. To overcome this limitation, DNA recombination systems such as Cre/loxP of bacteriophage P1, have been adapted for use in mammalian cells. The mutagenic event is restricted to the tissue or cell type in question leaving other body systems undisturbed. Conditional inactivation of Csk or Socs3, for example, established their key role in the prevention of inappropriate inflammation, while unexpected immunoregulatory activities emerged from studies of the NF-kappaB and AP-1 pathways. Also, cell types responsible for protective or pathogenic TNFalpha production have been identified. Inactivation of immunoregulatory receptors in leukocyte subsets can provide robust experimental systems revealing the conceptual simplicity underlying the modulation of complex signaling pathways during homeostatic responses. As illustrated for TGF-beta receptor, such system-guided approaches can provide a comprehensive picture of the regulatory events driving in vivo phenotype and specific responses of primary cells. This in turn facilitates the identification of novel regulatory mechanisms, targets for therapeutic intervention and prediction of side effects. With the increasing evidence for a role of somatic mutations in a wider range of human diseases, conditional mouse models are set to play a continuing part in the identification of pathogenic mechanisms for restoration of normal cellular processes in diseases including cancer, inflammation and autoimmunity.
Collapse
Affiliation(s)
- J Roes
- Department of Immunology and Molecular Pathology, University College London, 46 Cleveland Street, London W1T 4JF, UK.
| |
Collapse
|
588
|
Montanez E, Piwko-Czuchra A, Bauer M, Li S, Yurchenco P, Fässler R. Analysis of integrin functions in peri-implantation embryos, hematopoietic system, and skin. Methods Enzymol 2007; 426:239-89. [PMID: 17697888 DOI: 10.1016/s0076-6879(07)26012-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion, permit traction forces important for cell migration, and cross-talk with growth factor receptors to regulate cell proliferation, cell survival, and cell differentiation. The plethora of functions explains their central role for development and disease. The progress in mouse genetics and the ease with which the mouse genome can be manipulated enormously contributed to our understanding of how integrins exert their functions at the molecular level. In the present chapter, we describe tests that are routinely used in our laboratory to investigate embryos, organs, and cells (peri-implantation embryos, hematopoietic system, epidermis, and hair follicles) that lack the expression of integrins or integrin-associated proteins.
Collapse
Affiliation(s)
- Eloi Montanez
- Max Planck Institute of Biochemistry, Department of Molecular Medicine, Martinsried, Germany
| | | | | | | | | | | |
Collapse
|
589
|
Coles MC, Veiga-Fernandes H, Foster KE, Norton T, Pagakis SN, Seddon B, Kioussis D. Role of T and NK cells and IL7/IL7r interactions during neonatal maturation of lymph nodes. Proc Natl Acad Sci U S A 2006; 103:13457-62. [PMID: 16938836 PMCID: PMC1569185 DOI: 10.1073/pnas.0604183103] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2006] [Indexed: 12/14/2022] Open
Abstract
Lymph node (LN) development depends on prenatal interactions occurring between LN inducer and LN organizer cells. We have distinguished defects in LN formation due to failure in embryonic development (aly/aly) from defects in postnatal maturation (Il2rgamma(-/-)Rag2(-/-)). Both mutant strains form normal primordial LNs with differing fate. In aly/aly mice, the LN primordium dissipates irreversibly late in gestation; in contrast, Il2rgamma(-/-)Rag2(-/-) LN anlage persists for a week after birth but disperses subsequently, a process reversible by neonatal transfer of WT IL7r(+) TCR(+) T or natural killer (NK) cells, suggesting a role for IL7/IL7r interactions. Thus, we reveal a unique stage of postnatal LN development during which mature lymphocytes and IL7/IL7r interactions may play an important role.
Collapse
Affiliation(s)
| | | | | | | | - Stamatis N. Pagakis
- Confocal Microscopy and Image Analysis Laboratory, National Institute for Medical Research, Mill Hill, London NW7 1AA, United Kingdom
| | | | | |
Collapse
|
590
|
Harrow F, Ortiz BD. The TCRalpha locus control region specifies thymic, but not peripheral, patterns of TCRalpha gene expression. THE JOURNAL OF IMMUNOLOGY 2006; 175:6659-67. [PMID: 16272321 DOI: 10.4049/jimmunol.175.10.6659] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The molecular mechanisms ensuring the ordered expression of TCR genes are critical for proper T cell development. The mouse TCR alpha-chain gene locus contains a cis-acting locus control region (LCR) that has been shown to direct integration site-independent, lymphoid organ-specific expression of transgenes in vivo. However, the fine cell type specificity and developmental timing of TCRalpha LCR activity are both still unknown. To address these questions, we established a transgenic reporter model of TCRalpha LCR function that allows for analysis of LCR activity in individual cells by the use of flow cytometry. In this study we report the activation of TCRalpha LCR activity at the CD4-CD8-CD25-CD44- stage of thymocyte development that coincides with the onset of endogenous TCRalpha gene rearrangement and expression. Surprisingly, TCRalpha LCR activity appears to decrease in peripheral T cells where TCRalpha mRNA is normally up-regulated. Furthermore, LCR-linked transgene activity is evident in gammadelta T cells and B cells. These data show that the LCR has all the elements required to reliably reproduce a developmentally correct TCRalpha-like expression pattern during thymic development and unexpectedly indicate that separate gene regulatory mechanisms are acting on the TCRalpha gene in peripheral T cells to ensure its high level and fine cell type-specific expression.
Collapse
Affiliation(s)
- Faith Harrow
- Department of Biological Sciences, City University of New York, Hunter College, New York, NY 10021, USA
| | | |
Collapse
|
591
|
Zhang DJ, Wang Q, Wei J, Baimukanova G, Buchholz F, Stewart AF, Mao X, Killeen N. Selective expression of the Cre recombinase in late-stage thymocytes using the distal promoter of the Lck gene. THE JOURNAL OF IMMUNOLOGY 2005; 174:6725-31. [PMID: 15905512 DOI: 10.4049/jimmunol.174.11.6725] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Transgenic mouse lines were generated that express the Cre recombinase under the control of the distal promoter of the mouse Lck gene. Cre recombination in four of these lines of transgenic mice was characterized at the single cell level using ROSA26-regulated loxP-Stop-loxP-betageo and loxP-Stop-loxP-YFP reporter mouse lines. Two of the lines showed T cell-restricted Cre recombination, whereas the other two also expressed Cre in B cells, NK cells, and monocytes. Cre recombination began at a late stage of T cell development (at or after up-regulation of the TCR during positive selection) in the two T cell-restricted lines. Lines of mice that express the Cre recombinase at late stages of thymocyte development are of value for determining the impact of mutations on T cell function in the absence of complicating effects on early thymocyte selection.
Collapse
Affiliation(s)
- Dong Ji Zhang
- Department of Microbiology and Immunology, University of California, San Francisco, 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
592
|
Ferron M, Vacher J. Targeted expression of Cre recombinase in macrophages and osteoclasts in transgenic mice. Genesis 2005; 41:138-45. [PMID: 15754380 DOI: 10.1002/gene.20108] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To develop specific conditional gene ablation in the hematopoietic myeloid-osteoclast lineage, transgenic mice expressing Cre recombinase under the control of the CD11b promotor were generated on the C57BL/6 background. The cellular specificity of Cre activity following recombination was quantified in the Z/EG reporter transgenic mice by FACS analysis with lineage-specific markers and EGFP coexpression. A high degree of recombination, as evidenced by EGFP-positive cells, was demonstrated in macrophages and granulocytes of bone marrow and spleen by the presence of double-positive cells CD11b/EGFP and Gr1/EGFP, respectively. Interestingly, the peritoneal macrophage population showed almost complete DNA recombination at large. Most important, mature osteoclast cells derived from the double transgenic bone marrow and spleen progenitors were EGFP-positive. Hence, these CD11b-Cre mice will provide a unique tool to unravel novel gene function and activities involved during osteoclast and macrophage differentiation and maturation processes.
Collapse
Affiliation(s)
- M Ferron
- Institut de Recherches Cliniques de Montréal, Québec H2W 1R7, Canada
| | | |
Collapse
|
593
|
Bourgeois C, Kassiotis G, Stockinger B. A major role for memory CD4 T cells in the control of lymphopenia-induced proliferation of naive CD4 T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:5316-23. [PMID: 15843528 DOI: 10.4049/jimmunol.174.9.5316] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a state of lymphopenia, naive and memory CD4 T cells compete with each other for expansion at the expense of naive T cells. This competition prevents the proliferation as well as the phenotypic and functional conversion of naive T cells to "memory-like" T cells and may consequently prevent immune pathology frequently associated with lymphopenia-induced proliferation of naive cells. However, in T cell replete mice, memory T cells do not compete with naive T cells, indicating independent homeostatic control of naive and memory CD4 T cells in conditions that do not involve profound lymphopenia. Moreover, within the memory compartment, subsequent generation of new memory T cells precludes the survival of memory-like T cells. Thus, memory T cells have a major role in the control of lymphopenia-induced proliferation of naive cells because they inhibit both the generation of memory-like T cells and their persistence within the memory compartment.
Collapse
Affiliation(s)
- Christine Bourgeois
- Division of Molecular Immunology, National Institute for Medical Research, London, United Kingdom
| | | | | |
Collapse
|
594
|
Belteki G, Haigh J, Kabacs N, Haigh K, Sison K, Costantini F, Whitsett J, Quaggin SE, Nagy A. Conditional and inducible transgene expression in mice through the combinatorial use of Cre-mediated recombination and tetracycline induction. Nucleic Acids Res 2005; 33:e51. [PMID: 15784609 PMCID: PMC1069131 DOI: 10.1093/nar/gni051] [Citation(s) in RCA: 301] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Here we describe a triple transgenic mouse system, which combines the tissue specificity of any Cre-transgenic line with the inducibility of the reverse tetracycline transactivator (rtTA)/tetracycline-responsive element (tet-O)-driven transgenes. To ensure reliable rtTA expression in a broad range of cell types, we have targeted the rtTA transgene into the ROSA26 locus. The rtTA expression, however, is conditional to a Cre recombinase-mediated excision of a STOP region from the ROSA26 locus. We demonstrate the utility of this technology through the inducible expression of the vascular endothelial growth factor (VEGF-A) during embryonic development and postnatally in adult mice. Our results of adult induction recapitulate several different hepatic and immune cell pathological phenotypes associated with increased systemic VEGF-A protein levels. This system will be useful for studying genes in which temporal control of expression is necessary for the discovery of the full spectrum of functions. The presented approach abrogates the need to generate tissue-specific rtTA transgenes for tissues where well-characterized Cre lines already exist.
Collapse
Affiliation(s)
- Gusztav Belteki
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Jody Haigh
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Nikolett Kabacs
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Katharina Haigh
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Karen Sison
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Frank Costantini
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia UniversityNew York, NY 10032, USA
| | - Jeff Whitsett
- Children's Hospital Medical Center, Division of Pulmonary BiologyCincinnati, OH 45229-3039, USA
| | - Susan E. Quaggin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
- St Michael's HospitalToronto, Ontario, Canada
| | - Andras Nagy
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital600 University Avenue, Toronto, Ontario, Canada M5G 1X5
- Department of Molecular and Medical Genetics, University of TorontoToronto, Ontario, Canada M5S 1A8
- To whom correspondence should be addressed. Tel: +1 416 586 3246; Fax: +1 416 586 8588;
| |
Collapse
|
595
|
Kassiotis G, Stockinger B. Anatomical Heterogeneity of Memory CD4+ T Cells Due to Reversible Adaptation to the Microenvironment. THE JOURNAL OF IMMUNOLOGY 2004; 173:7292-8. [PMID: 15585852 DOI: 10.4049/jimmunol.173.12.7292] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The memory T cell pool is characterized by a substantial degree of heterogeneity in phenotype and function as well as anatomical distribution, but the underlying mechanisms remain unclear. In this study we confirm that the memory CD4(+) T cell pool in wild-type and TCR-transgenic mice consists of heterogeneous subsets, as defined by surface marker expression or cytokine production. Extralymphoid sites contain significant numbers of memory CD4(+) T cells, which are phenotypically and functionally distinct from their lymphoid counterparts. However, we show in this study that the phenotype of lymphoid and extralymphoid memory T cells is not stable. Instead, the unique properties of extralymphoid memory T cells are acquired upon migration into extralymphoid sites and are lost when memory T cells migrate back into lymphoid organs. Thus, at least some of the extralymphoid properties may represent a transient activation state that can be adopted by T cells belonging to a single memory T cell pool. Furthermore, such intermittent activation during or after migration into extralymphoid sites could provide an important signal, promoting the survival and functional competence of memory T cells in the absence of Ag.
Collapse
Affiliation(s)
- George Kassiotis
- Division of Molecular Immunology, National Institute for Medical Research, London, United Kingdom
| | | |
Collapse
|
596
|
Stadtfeld M, Graf T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development 2004; 132:203-13. [PMID: 15576407 DOI: 10.1242/dev.01558] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Hematopoietic cells have been reported to convert into a number of non-hematopoietic cells types after transplantation/injury. Here, we have used a lineage tracing approach to determine whether hematopoietic plasticity is relevant for the normal development of hepatocytes and endothelial cells, both of which develop in close association with blood cells. Two mouse models were analyzed: vav ancestry mice, in which essentially all hematopoietic cells, including stem cells, irreversibly express yellow fluorescent protein (YFP); and lysozyme ancestry mice, in which all macrophages, as well as a small subset of all other non-myeloid hematopoietic cells, are labeled. Both lines were found to contain YFP+ hepatocytes at similar frequencies, indicating that macrophage to hepatocyte contributions occur in unperturbed mice. However, the YFP+ hepatocytes never formed clusters larger than three cells, suggesting a postnatal origin. In addition, the frequency of these cells was very low (approximately 1 in 75,000) and only increased two- to threefold after acute liver injury. Analysis of the two mouse models revealed no evidence for a hematopoietic origin of endothelial cells, showing that definitive HSCs do not function as hemangioblasts during normal development. Using endothelial cells and hepatocytes as paradigms, our study indicates that hematopoietic cells are tightly restricted in their differentiation potential during mouse embryo development and that hematopoietic plasticity plays at best a minor role in adult organ maintenance and regeneration.
Collapse
Affiliation(s)
- Matthias Stadtfeld
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | |
Collapse
|
597
|
De Jersey J, Carmignac D, Le Tissier P, Barthlott T, Robinson I, Stockinger B. Factors affecting the susceptibility of the mouse pituitary gland to CD8 T-cell-mediated autoimmunity. Immunology 2004; 111:254-61. [PMID: 15009425 PMCID: PMC1782418 DOI: 10.1111/j.1365-2567.2004.01821.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We have previously shown, in a transgenic mouse model, that the pituitary gland is susceptible to CD8 T-cell-mediated autoimmunity, triggered by a cell-specific model autoantigen, resulting in pan-anterior pituitary hypophysitis and dwarfism. In the present study, we now demonstrate that antigen dose, the T-cell precursor frequency, the degree of lymphopenia and the context of target antigen expression, are important parameters determining the time course and extent of the pathological consequences of CD8 T-cell-mediated autoimmunity. Furthermore, our data indicate that the pituitary gland is susceptible to CD8 autoimmunity following an inflammatory insult such as a viral infection. As lymphocytic hypophysitis may be manifest in other autoimmune conditions, and the pituitary gland may be susceptible to T-cell-mediated pathology after immunization with a virus expressing soluble pituitary antigen, it is important to consider that strategies based on vaccination against soluble pituitary gonadotrophins could have other unexpected endocrine consequences.
Collapse
Affiliation(s)
- James De Jersey
- Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | | | |
Collapse
|
598
|
Almarza E, Segovia JC, Guenechea G, Gómez SG, Ramírez A, Bueren JA. Regulatory elements of the vav gene drive transgene expression in hematopoietic stem cells from adult mice. Exp Hematol 2004; 32:360-4. [PMID: 15050746 DOI: 10.1016/j.exphem.2004.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2003] [Revised: 11/25/2003] [Accepted: 01/16/2004] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Previous studies have shown that the HS21/45 promoter of the vav protooncogene drives a predominant expression of exogenous transgenes in mouse hematopoietic cells, including clonogenic bone marrow (BM) progenitors. We investigated the activity of this promoter in the hematopoietic stem cell compartment of adult mice. MATERIALS AND METHODS Inbred Ly5.1 transgenic mice expressing a nonfunctional human CD4 marker gene (hCD4) under the control of the HS21/45 promoter were generated. BM cells from these animals were sorted based on the intensity of hCD4 expression. Fractions characterized by high, intermediate, or low/negative expression of the transgene were then assessed for their competitive repopulation ability (CRA), using unfractionated BM cells from Ly5.2 mice as a reference competitor population. RESULTS Data showed that BM cells having a low/negative or intermediate expression of hCD4 had a very poor hematopoietic CRA. In contrast, BM cells with high hCD4 expression were characterized by a high CRA. These observations were confirmed in the short- and long-term posttransplantation of primary and secondary recipients when analyzing the lymphoid and myeloid cells of recipient mice. CONCLUSIONS Our results demonstrate for the first time that the regulatory HS21/45 sequence of the vav gene constitutes an efficient promoter for driving transgene expression in multipotent hematopoietic stem cells residing in the BM of adult mice. Thus, this promoter is proposed for the development of transgenic mice and gene therapy vectors that require restricted expression of exogenous transgenes in cells of the hematopoietic system, including primitive hematopoietic stem cells.
Collapse
Affiliation(s)
- Elena Almarza
- Gene Therapy Programme, Centro de Investigaciones, Energéticas, Medioambientales y Tecnológicas/Marcelino Botín Foundation, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
599
|
Croker BA, Metcalf D, Robb L, Wei W, Mifsud S, DiRago L, Cluse LA, Sutherland KD, Hartley L, Williams E, Zhang JG, Hilton DJ, Nicola NA, Alexander WS, Roberts AW. SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity 2004; 20:153-65. [PMID: 14975238 DOI: 10.1016/s1074-7613(04)00022-6] [Citation(s) in RCA: 219] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2003] [Revised: 12/18/2003] [Accepted: 12/22/2003] [Indexed: 11/20/2022]
Abstract
To determine the importance of suppressor of cytokine signaling-3 (SOCS3) in the regulation of hematopoietic growth factor signaling generally, and of G-CSF-induced cellular responses specifically, we created mice in which the Socs3 gene was deleted in all hematopoietic cells. Although normal until young adulthood, these mice then developed neutrophilia and a spectrum of inflammatory pathologies. When stimulated with G-CSF in vitro, SOCS3-deficient cells of the neutrophilic granulocyte lineage exhibited prolonged STAT3 activation and enhanced cellular responses to G-CSF, including an increase in cloning frequency, survival, and proliferative capacity. Consistent with the in vitro findings, mutant mice injected with G-CSF displayed enhanced neutrophilia, progenitor cell mobilization, and splenomegaly, but unexpectedly also developed inflammatory neutrophil infiltration into multiple tissues and consequent hind-leg paresis. We conclude that SOCS3 is a key negative regulator of G-CSF signaling in myeloid cells and that this is of particular significance during G-CSF-driven emergency granulopoiesis.
Collapse
Affiliation(s)
- Ben A Croker
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research and The Cooperative Research Centre for Cellular Growth Factors, Parkville, Victoria 3050, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
600
|
Yannoutsos N, Barreto V, Misulovin Z, Gazumyan A, Yu W, Rajewsky N, Peixoto BR, Eisenreich T, Nussenzweig MC. A cis element in the recombination activating gene locus regulates gene expression by counteracting a distant silencer. Nat Immunol 2004; 5:443-50. [PMID: 15021880 DOI: 10.1038/ni1053] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2004] [Accepted: 01/22/2004] [Indexed: 12/22/2022]
Abstract
We have identified a silencer and an antisilencing element that interact at a distance of 85 kilobases to regulate expression of the recombination activating genes Rag1 and Rag2 in thymocytes. Transgenic experiments showed that Rag promoter-proximal cis elements directed tissue-specific expression and that a Runx-dependent intergenic silencer suppressed expression in developing T cells. Deletion of the antisilencing element from the genomic Rag locus unmasked the intergenic silencer and abrogated Rag expression in developing CD4(+)CD8(+) T cells. We speculate that the Rag antisilencing element belongs to a class of cis elements that might be useful for genome diversification by activating genes encoded by otherwise silent transposable elements.
Collapse
Affiliation(s)
- Nikos Yannoutsos
- Laboratory of Molecular Immunology, Rockefeller University, New York, New York 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|