2601
|
Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, Cumano A, Lauvau G, Geissmann F. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 2007; 317:666-70. [PMID: 17673663 DOI: 10.1126/science.1142883] [Citation(s) in RCA: 1456] [Impact Index Per Article: 80.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cellular immune response to tissue damage and infection requires the recruitment of blood leukocytes. This process is mediated through a classical multistep mechanism, which involves transient rolling on the endothelium and recognition of inflammation followed by extravasation. We have shown, by direct examination of blood monocyte functions in vivo, that a subset of monocytes patrols healthy tissues through long-range crawling on the resting endothelium. This patrolling behavior depended on the integrin LFA-1 and the chemokine receptor CX(3)CR1 and was required for rapid tissue invasion at the site of an infection by this "resident" monocyte population, which initiated an early immune response and differentiated into macrophages.
Collapse
Affiliation(s)
- Cedric Auffray
- Institut Nationale de la Santé et de la Recherche Médicale (INSERM) U838, Laboratory of Biology of the Mononuclear Phagocyte System, and Cellular and Molecular imaging core facility, Institut Fédératif de Recherche Necker-Enfants Malades, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2602
|
Villadangos JA, Schnorrer P. Intrinsic and cooperative antigen-presenting functions of dendritic-cell subsets in vivo. Nat Rev Immunol 2007; 7:543-55. [PMID: 17589544 DOI: 10.1038/nri2103] [Citation(s) in RCA: 480] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) comprise several subsets, and their roles in the presentation of antigens derived from pathogens, vaccines and self tissues are now beginning to be elucidated. Differences in location, life cycle and intrinsic abilities to capture, process and present antigens on their MHC class I and class II molecules enable each DC subset to have distinct roles in immunity to infection and in the maintenance of self tolerance. Unexpected interactions among DC subsets have also been revealed. These interactions, which allow the integration of the intrinsic abilities of different DC types, enhance the ability of the DC network to respond to multiple scenarios of infection.
Collapse
Affiliation(s)
- José A Villadangos
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | |
Collapse
|
2603
|
Abstract
Dendritic cells (DCs) are a heterogenous population of bone-marrow-derived immune cells. Although all DCs share a common ability to process and present antigen to naive T cells for the initiation of an immune response, they differ in surface markers, migratory patterns, localization, and cytokine production. DCs were originally considered to be myeloid cells, but recent findings have demonstrated that DCs can develop not only from myeloid- but also from lymphoid-committed progenitors. The common feature of the progenitors capable of developing into DCs is the surface expression of Flt3 receptor. The development of different populations of DCs is differentially regulated by various transcription factors and cytokines. This review summarizes the recent advances made in the field of DC development.
Collapse
Affiliation(s)
- Li Wu
- Immunology Division, The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | |
Collapse
|
2604
|
Abstract
The role of macrophages in modulating the systemic response to hypoxia and oxidative stress is emerging from basic biological processes, such as the regulation of red blood cell production, and from analysis of tumor progression, as a key factor determining whether cells survive, proliferate or differentiate under micro-environmental pressures. Our recent work identified a novel role for macrophages in promoting expansion of erythroid progenitors in vitro while confirming previous data that macrophages are not required for red cell enucleation. This work emerged from analyses of hypoxia and cell death in the Rb null fetal liver where we demonstrated that defects in erythropoietic islands were due to deterioration in the fetal liver microenvironment that disrupted heterotypic interactions of macrophages with erythroblasts and not to intrinsic defects in Rb null macrophages. The significance of these findings for the effect of hypoxia on macrophage interactions and activity during tumor progression is also discussed.
Collapse
Affiliation(s)
| | - Kay F. Macleod
- Correspondence to: Kay Macleod; The Ben May Department for Cancer Research; The University of Chicago; GCIS-W338; 929 East 57th Street; Chicago, Illinois 60637 USA;
| |
Collapse
|
2605
|
Wick MJ. Monocyte and dendritic cell recruitment and activation during oral Salmonella infection. Immunol Lett 2007; 112:68-74. [PMID: 17720254 DOI: 10.1016/j.imlet.2007.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 07/13/2007] [Accepted: 07/15/2007] [Indexed: 12/24/2022]
Abstract
Immunity to bacterial infection involves the joint effort of the innate and adaptive immune systems. The innate immune response is triggered when the body senses bacterial components, such as lipopolysaccharide, that alarm the body of the invader. An array of cell types function in the innate response. These cells are rapidly recruited to the infection site and activated to optimally perform their functions. The adaptive immune response follows the innate response, and one cell type in particular, dendritic cells (DCs), are the critical link between the innate and adaptive responses. This review will summarize recent data concerning the events that occur early during oral infection with the intracellular pathogen Salmonella, with emphasis on the phagocytic cells involved in combating the infection in the gut-associated lymphoid tissues. In particular, recent findings concerning the recruitment and activation of mononuclear phagocyte populations and dendritic cell subsets will be presented after an overview of the Salmonella infection model.
Collapse
Affiliation(s)
- Mary Jo Wick
- Department of Microbiology and Immunology, Göteborg University, Box 435, S-405 30 Göteborg, Sweden.
| |
Collapse
|
2606
|
Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol 2007; 8:942-9. [PMID: 17676045 DOI: 10.1038/ni1496] [Citation(s) in RCA: 1562] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 06/26/2007] [Indexed: 11/08/2022]
Abstract
Interleukin 17 (IL-17)-producing CD4(+) helper T cells (T(H)-17 cells) have been linked to host defense and autoimmune diseases. In mice, the differentiation of T(H)-17 cells requires transforming growth factor-beta and IL-6 and the transcription factor RORgammat. We report here that for human naive CD4(+) T cells, RORgammat expression and T(H)-17 polarization were induced by IL-1beta and enhanced by IL-6 but were suppressed by transforming growth factor-beta and IL-12. Monocytes and conventional dendritic cells, but not monocyte-derived dendritic cells activated by microbial stimuli, efficiently induced T(H)-17 priming, and this function correlated with antigen-presenting cell production of IL-1beta and IL-6 but not IL-12. Our results identify cytokines, antigen-presenting cells and microbial products that promote the polarization of human T(H)-17 cells and emphasize an important difference in the requirements for the differentiation of T(H)-17 cells in humans and mice.
Collapse
|
2607
|
Rae F, Woods K, Sasmono T, Campanale N, Taylor D, Ovchinnikov DA, Grimmond SM, Hume DA, Ricardo SD, Little MH. Characterisation and trophic functions of murine embryonic macrophages based upon the use of a Csf1r–EGFP transgene reporter. Dev Biol 2007; 308:232-46. [PMID: 17597598 DOI: 10.1016/j.ydbio.2007.05.027] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 05/08/2007] [Accepted: 05/22/2007] [Indexed: 11/20/2022]
Abstract
All solid organs contain resident monocyte-derived cells that appear early in organogenesis and persist throughout life. These cells are critical for normal development in some organs. Here we report the use of a previously described transgenic line, with EGFP driven by the macrophage-restricted Csf1r (c-fms) promoter, to image macrophage production and infiltration accompanying organogenesis in many tissues. Using microarray analysis of FACS-isolated EGFP-positive cells, we show that fetal kidney, lung and brain macrophages show similar gene expression profiles irrespective of their tissue of origin. EGFP-positive cells appeared in the renal interstitium from 12 days post coitum, prior to nephrogenesis, and maintain a close apposition to renal tubules postnatally. CSF-1 added to embryonic kidney explants increased overall renal growth and ureteric bud branching. Expression profiling of tissue macrophages and of CSF-1-treated explants showed evidence of the alternate, pro-proliferative (M2) activation profile, including expression of macrophage mannose receptor (CD206), macrophage scavenger receptor 2 (Msr2), C1q, CD163, selenoprotein P, CCL24 and TREM2. This response has been associated with the trophic role of tumour-associated macrophages. These findings suggest a trophic role of macrophages in embryonic kidney development, which may continue to play a similar role in postnatal repair.
Collapse
Affiliation(s)
- Fiona Rae
- Institute for Molecular Bioscience and ARC Special Research Centre for Functional and Applied Genomics, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2608
|
Nalubamba KS, Gossner AG, Dalziel RG, Hopkins J. Differential expression of pattern recognition receptors in sheep tissues and leukocyte subsets. Vet Immunol Immunopathol 2007; 118:252-62. [PMID: 17604125 DOI: 10.1016/j.vetimm.2007.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Revised: 05/30/2007] [Accepted: 05/31/2007] [Indexed: 01/30/2023]
Abstract
The various members of the different pattern recognition receptor families are now recognized as playing a crucial role in the initial interactions between a pathogen and the host. This paper identifies all 10 members of the TLR family in sheep as well as key members of the C-type lectin and NLR families. Our data show that sheep possess the 'human' and not the 'mouse' pattern of TLRs and confirm the high degree of sequence identity between orthologous genes in the different species. In the absence of definitive antibodies, qRT-PCR assays were developed to quantify PRR transcript expression patterns in a range of normal sheep tissues as well as isolated dendritic cell (DC) and leukocyte subsets. These data show that the lymphoid organs (spleen and lymph nodes) express the widest range of PRRs and that organs such as the lung and kidney have distinctive arrays of PRRs that reflect their potential risk of pathogen exposure. In addition we show that the two DC subsets, defined by the differential expression of CD172a/CD45RA and their cytokine expression profiles, have different and characteristic PRR complements again possibly reflecting their distinctive function. These data are important for future studies on the role of PRRs in disease pathogenesis and control.
Collapse
Affiliation(s)
- King S Nalubamba
- Centre of Infectious Diseases, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Summerhall, Edinburgh EH9 1QH, UK
| | | | | | | |
Collapse
|
2609
|
Tajima K, Terai S, Takami T, Kawaguchi K, Okita K, Sakaida I. Importance of inhibitor of DNA binding/differentiation 2 in hepatic stellate cell differentiation and proliferation. Hepatol Res 2007; 37:647-55. [PMID: 17559421 DOI: 10.1111/j.1872-034x.2007.00089.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIM In liver fibrosis, activated hepatic stellate cells (HSC) are transformed into myofibroblasts. Helix-loop-helix (HLH) transcriptional factors such as MyoD regulate the differentiation of myocytes, and the inhibitor of DNA binding/differentiation (Id) family comprises dominant negative HLH transcriptional regulators that inhibit differentiation and promote cell proliferation. In the present study, we investigated how the Id family proteins regulate HSC. METHODS In primary rat HSC, inhibitor of DNA binding/differentiation (Id)2 and alpha-smooth muscle actin (alpha-SMA) mRNA expression increased 4 days after isolation. Next we established Id2 expressing HSC (HSC-T6-Id2-green fluorescent protein (GFP)) using HSC-T6 cells with retrovirus that expressed GFP-tagged Id2. RESULTS HSC-T6-Id2-GFP increased cell proliferation with cyclin D1 expression. In contrast, alpha-SMA expression wassuppressed. Real-time reverse transcription-polymerase chain reaction analysis showed Id2 induction significantly suppressed alpha-SMA, collagen-1, matrix metalloproteinase (MMP)-2, and MMP-9 mRNA (P < 0.05) but had no effect on tissue inhibitor of metalloproteinase or transforming growth factor-beta1 levels. CONCLUSION These findings suggest Id2, an HLH transcriptional regulator, plays an important regulatory role in the proliferation and differentiation of HSC.
Collapse
Affiliation(s)
- Kunihiko Tajima
- Department of Molecular Science and Applied Medicine (Gastroenterology and Hepatology), Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
2610
|
|
2611
|
Iqbal J, Greiner TC, Patel K, Dave BJ, Smith L, Ji J, Wright G, Sanger WG, Pickering DL, Jain S, Horsman DE, Shen Y, Fu K, Weisenburger DD, Hans CP, Campo E, Gascoyne RD, Rosenwald A, Jaffe ES, Delabie J, Rimsza L, Ott G, Müller-Hermelink HK, Connors JM, Vose JM, McKeithan T, Staudt LM, Chan WC. Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma. Leukemia 2007; 21:2332-43. [PMID: 17625604 PMCID: PMC2366166 DOI: 10.1038/sj.leu.2404856] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gene expression profiling of diffuse large B-cell lymphoma (DLBCL) has revealed biologically and prognostically distinct subgroups: germinal center B-cell-like (GCB), activated B-cell-like (ABC) and primary mediastinal (PM) DLBCL. The BCL6 gene is often translocated and/or mutated in DLBCL. Therefore, we examined the BCL6 molecular alterations in these DLBCL subgroups, and their impact on BCL6 expression and BCL6 target gene repression. BCL6 translocations at the major breakpoint region (MBR) were detected in 25 (18.8%) of 133 DLBCL cases, with a higher frequency in the PM (33%) and ABC (24%) subgroups than in the GCB (10%) subgroup. Translocations at the alternative breakpoint region (ABR) were detected in five (6.4%) of 78 DLBCL cases, with three cases in ABC and one case each in the GCB and the unclassifiable subgroups. The translocated cases involved IgH and non-IgH partners in about equal frequency and were not associated with different levels of BCL6 mRNA and protein expression. BCL6 mutations were detected in 61% of DLBCL cases, with a significantly higher frequency in the GCB and PM subgroups (>70%) than in the ABC subgroup (44%). Exon-1 mutations were mostly observed in the GCB subgroup. The repression of known BCL6 target genes correlated with the level of BCL6 mRNA and protein expression in GCB and ABC subgroups but not with BCL6 translocation and intronic mutations. No clear inverse correlation between BCL6 expression and p53 expression was observed. Patients with higher BCL6 mRNA or protein expression had a significantly better overall survival. The biological role of BCL6 in translocated cases where repression of known target genes is not demonstrated is intriguing and warrants further investigation.
Collapse
Affiliation(s)
- J Iqbal
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - TC Greiner
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Patel
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - BJ Dave
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - L Smith
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - J Ji
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - G Wright
- Metabolism Branch and Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - WG Sanger
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - DL Pickering
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Jain
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - DE Horsman
- Departments of Pathology and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Y Shen
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Fu
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - DD Weisenburger
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - CP Hans
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - E Campo
- Department of Pathology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - RD Gascoyne
- Departments of Pathology and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - A Rosenwald
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | - ES Jaffe
- Metabolism Branch and Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Delabie
- Norwegian Radium Hospital, Oslo, Norway
| | - L Rimsza
- Department of Pathology, University of Arizona, Tucson, Arizona, USA
| | - G Ott
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | | | - JM Connors
- Departments of Pathology and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - JM Vose
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - T McKeithan
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - LM Staudt
- Metabolism Branch and Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - WC Chan
- Departments of Pathology and Microbiology, Pediatrics, Internal Medicine, and Preventive and Societal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
2612
|
Merad M, Collin M, Bromberg J. Dendritic cell homeostasis and trafficking in transplantation. Trends Immunol 2007; 28:353-9. [PMID: 17618832 DOI: 10.1016/j.it.2007.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 05/22/2007] [Accepted: 06/20/2007] [Indexed: 10/23/2022]
Abstract
Hematopoietic cell transplantation and solid organ transplantation are definitive therapies for several otherwise fatal conditions. Post-transplant immune reactions are the major cause of morbidity after transplantation and limit the extended use of these critical therapies. Post-transplant immune complications include graft rejection by the host and injury to the host mediated by the graft. Dendritic cells (DCs), a population of professional antigen-presenting cells, are thought to be crucial in triggering primary immune responses against both the graft and the host. Here, we review studies on DC homeostasis and trafficking after transplantation, and examine the role of the host and graft DC in post-transplant immune responses. We also discuss the therapeutic implications of these studies.
Collapse
Affiliation(s)
- Miriam Merad
- Department of Gene and Cell Medicine, Mount Sinai Medical School, 1425 Madison Avenue, New York, NY 10029, USA.
| | | | | |
Collapse
|
2613
|
Bajaña S, Herrera-González N, Narváez J, Torres-Aguilar H, Rivas-Carvalho A, Aguilar SR, Sánchez-Torres C. Differential CD4(+) T-cell memory responses induced by two subsets of human monocyte-derived dendritic cells. Immunology 2007; 122:381-93. [PMID: 17608690 PMCID: PMC2266029 DOI: 10.1111/j.1365-2567.2007.02650.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Dendritic cells (DC) are powerful inducers of primary T-cell responses, but their role in secondary responses has not been extensively analysed. Here, we address the role of two DC subsets derived from human CD16(+) (16(+) mDC) or CD16(-) (16(-) mDC) monocytes on the reactivation of memory responses. CD4(+) CD45RA(-) memory T cells were obtained from adult blood donors, and central (T(CM)) and effector (T(EM)) memory T cells were isolated by fluorescence-activated cell sorting with anti-CCR7 antibodies. The 16(+) mDC and 16(-) mDC were cocultured with autologous lymphocytes, either unpulsed or loaded with purified protein derivatives of Mycobacterium tuberculosis (PPD) or tetanus toxoid (TT), and were analysed for up to 8 days. Over a range of doses, 16(+) mDC drove stronger T-cell proliferative responses against both antigens. Overall, antigen-specific memory cells tended to acquire a phenotype of T(EM) at later time-points in the culture, whereas cells that had completed fewer cycles of division were similar to T(CM). The 16(+) mDC induced higher rates of proliferation on both T(CM) and T(EM) lymphocytes than 16(-) mDC. This phenomenon was not related to the ability of both DC to induce CD25 expression on T cells, to lower secretion of interleukin-2, or to raise production of interleukin-10 during T-cell/16(-) mDC cocultures. The induction of T(CM) effector capacity in terms of interferon-gamma production was faster and more pronounced with 16(+) mDC, whereas both DC had similar abilities with T(EM). In conclusion, these data might reveal new potentials in vaccination protocols with 16(+) mDC aimed at inducing strong responses on central memory T cells.
Collapse
Affiliation(s)
- Sandra Bajaña
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Mexico City, Mexico
| | | | | | | | | | | | | |
Collapse
|
2614
|
Rydström A, Wick MJ. Monocyte recruitment, activation, and function in the gut-associated lymphoid tissue during oral Salmonella infection. THE JOURNAL OF IMMUNOLOGY 2007; 178:5789-801. [PMID: 17442963 DOI: 10.4049/jimmunol.178.9.5789] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neutrophils, monocytes, and dendritic cells (DC) are phenotypically and functionally related phagocytes whose presence in infected tissues is critical to host survival. Their overlapping expression pattern of surface molecules, the differentiation capacity of monocytes, and the presence of monocyte subsets underscores the complexity of understanding the role of these cells during infection. In this study we use five- to seven-color flow cytometry to assess the phenotype and function of monocytes recruited to Peyer's patches (PP) and mesenteric lymph nodes (MLN) after oral Salmonella infection of mice. The data show that CD68(high)Gr-1(int) (intermediate) monocytes, along with CD68(int)Gr-1(high) neutrophils, rapidly accumulate in PP and MLN. The monocytes have increased MHC-II and costimulatory molecule expression and, in contrast to neutrophils and DC, produce inducible NO synthase. Although neutrophils and monocytes from infected mice produce TNF-alpha and IL-1beta upon ex vivo culture, DC do not. In addition, although recruited monocytes internalize Salmonella in vitro and in vivo they did not induce the proliferation of OT-II CD4(+) T cells after coincubation with Salmonella expressing OVA despite their ability to activate OT-II cells when pulsed with the OVA(323-339) peptide. We also show that recruited monocytes enter the PP of infected mice independently of the mucosal address in cell adhesion molecule-1 (MAdCAM-1). Finally, recruited but not resident monocytes increase in the blood of orally infected mice, and MHC-II up-regulation, but not TNF-alpha or iNOS production, occur already in the blood. These studies are the first to describe the accumulation and function of monocyte subsets in the blood and GALT during oral Salmonella infection.
Collapse
Affiliation(s)
- Anna Rydström
- Department of Microbiology and Immunology, Göteborg University, Göteborg, Sweden
| | | |
Collapse
|
2615
|
Hibbs ML, Quilici C, Kountouri N, Seymour JF, Armes JE, Burgess AW, Dunn AR. Mice lacking three myeloid colony-stimulating factors (G-CSF, GM-CSF, and M-CSF) still produce macrophages and granulocytes and mount an inflammatory response in a sterile model of peritonitis. THE JOURNAL OF IMMUNOLOGY 2007; 178:6435-43. [PMID: 17475873 DOI: 10.4049/jimmunol.178.10.6435] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
To assess the combined role of G-CSF, GM-CSF, and M-CSF in myeloid cell production, mice deficient in all three myeloid CSFs were generated (G-/-GM-/-M-/- mice). G-/-GM-/-M-/- mice share characteristics found in mice lacking individual cytokines: they are toothless and osteopetrotic and furthermore acquire alveolar proteinosis that is more severe than that found in either GM-/- or G-/-GM-/- mice. G-/-GM-/-M-/- mice have a significantly reduced lifespan, which is prolonged by antibiotic administration, suggesting compromised ability to control bacterial infection. G-/-GM-/-M-/- mice have circulating neutrophils and monocytes, albeit at significantly reduced numbers compared with wild-type mice, but surprisingly, have more circulating monocytes than M-/- mice and more circulating neutrophils than G-/-GM-/- mice. Due to severe osteopetrosis, G-/-GM-/-M-/- mice show diminished numbers of myeloid cells, myeloid progenitors, and B lymphocytes in the bone marrow, but have significantly enhanced compensatory splenic hemopoiesis. Although G-/-GM-/-M-/- mice have a profound deficiency of myeloid cells in the resting peritoneal cavity, the animals mount a moderate cellular response in a model of sterile peritonitis. These data establish that in the absence of G-CSF, GM-CSF, and M-CSF, additional growth factor(s) can stimulate myelopoiesis and acute inflammatory responses.
Collapse
Affiliation(s)
- Margaret L Hibbs
- Signal Transduction Laboratory, Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Royal Melbourne Hospital, Victoria, and Department of Medicine, University of Melbourne, Parkville, Australia.
| | | | | | | | | | | | | |
Collapse
|
2616
|
Spike BT, Dibling BC, Macleod KF. Hypoxic stress underlies defects in erythroblast islands in the Rb-null mouse. Blood 2007; 110:2173-81. [PMID: 17557897 PMCID: PMC1976369 DOI: 10.1182/blood-2007-01-069104] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Definitive erythropoiesis occurs in islands composed of a central macrophage in contact with differentiating erythroblasts. Erythroid maturation including enucleation can also occur in the absence of macrophages both in vivo and in vitro. We reported previously that loss of Rb induces cell-autonomous defects in red cell maturation under stress conditions, while other reports have suggested that the failure of Rb-null erythroblasts to enucleate is due to defects in associated macrophages. Here we show that erythropoietic islands are disrupted by hypoxic stress, such as occurs in the Rb-null fetal liver, that Rb(-/-) macrophages are competent for erythropoietic island formation in the absence of exogenous stress and that enucleation defects persist in Rb-null erythroblasts irrespective of macrophage function.
Collapse
Affiliation(s)
- Benjamin T Spike
- Ben May Department for Cancer Research, Center for Integrative Sciences, Chicago, IL 60637, USA
| | | | | |
Collapse
|
2617
|
Shaffer AL, Wright G, Yang L, Powell J, Ngo V, Lamy L, Lam LT, Davis RE, Staudt LM. A library of gene expression signatures to illuminate normal and pathological lymphoid biology. Immunol Rev 2007; 210:67-85. [PMID: 16623765 DOI: 10.1111/j.0105-2896.2006.00373.x] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Genomics has provided a lever to pry open lymphoid cells and examine their regulatory biology. The large body of available gene expression data has also allowed us to define the of coordinately expressed genes, termed gene expression signatures, which characterize the states of cellular physiology that reflect cellular differentiation, activation of signaling pathways, and the action of transcription factors. Gene expression signatures that reflect the action of individual transcription factors can be defined by perturbing transcription factor function using RNA interference (RNAi), small-molecule inhibition, and dominant-negative approaches. We have used this methodology to define gene expression signatures of various transcription factors controlling B-cell differentiation and activation, including BCL-6, B lymphocyte-induced maturation protein-1 (Blimp-1), X-box binding protein-1 (XBP1), nuclear factor-kappaB (NF-kappaB), and c-myc. We have also curated a wide variety of gene expression signatures from the literature and assembled these into a signature database. Statistical methods can define whether any signature in this database is differentially expressed in independent biological samples, an approach we have used to gain mechanistic insights into the origin and clinical behavior of B-cell lymphomas. We also discuss the use of genomic-scale RNAi libraries to identify genes and pathways that may serve as therapeutic targets in B-cell malignancies.
Collapse
Affiliation(s)
- Arthur L Shaffer
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
2618
|
Abstract
Osteoclasts are cells of monocyte/macrophage origin that degrade bone matrix. Receptor activator of NF-kappaB ligand (RANKL) induces osteoclast differentiation in the presence of macrophage colony-stimulating factor. RANKL activates the tumor necrosis factor receptor-associated factor 6, c-Fos, and calcium signaling pathways, all of which are indispensable for the induction and activation of nuclear factor of activated T cells (NFAT) c1. NFATc1 is the master transcription factor for osteoclast differentiation, which regulates many osteoclast-specific genes. Multiple immunoglobulin-like receptors associated with immunoreceptor tyrosine-based activation motif (ITAM)-harboring adapters, Fc receptor common chi subunit (FcRgamma), and DNAX-activating protein (DAP) 12 mediate costimulatory signals for RANK, which activate calcium signaling through phospholipase Cgamma (PLCgamma). In addition to calcineurin-NFATc1, calcium signaling activates the CaMK-CREB (calcium/calmodulin activated kinase-cyclic AMP-response element binding protein) pathway, which also plays a critical role in osteoclastogenesis. This review summarizes recent advances in the study of signaling mechanisms of osteoclast differentiation.
Collapse
Affiliation(s)
- Masahiro Shinohara
- Department of Cell Signaling, Graduate School, Tokyo Medical and Dental University and COE Program for Frontier Research on Molecular Destruction and Reconstruction of Tooth and Bone, Yushima 1-5-45, Bunkyo-ku, Tokyo 113-8549, Japan.
| | | |
Collapse
|
2619
|
Pakuts B, Debonneville C, Liontos LM, Loreto MP, McGlade CJ. The Src-like Adaptor Protein 2 Regulates Colony-stimulating Factor-1 Receptor Signaling and Down-regulation. J Biol Chem 2007; 282:17953-17963. [PMID: 17353186 DOI: 10.1074/jbc.m701182200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Src-like adaptor protein 2 (SLAP-2) is a hematopoietic adaptor protein previously implicated as a negative regulator of T-cell antigen receptor (TCR)-mediated signaling. SLAP-2 contains an SH3 and an SH2 domain, followed by a unique carboxyl-terminal tail, which is important for c-Cbl binding. Here we describe a novel role for SLAP-2 in regulation of the colony-stimulating factor 1 receptor (CSF-1R), a receptor tyrosine kinase important for growth and differentiation of myeloid cells. SLAP-2 co-immunoprecipitates with c-Cbl and CSF-1R in primary bone marrow-derived macrophages. Using murine myeloid cells expressing CSF-1R (FD-Fms cells), we show that SLAP-2 is tyrosine-phosphorylated upon stimulation with CSF-1 and associates constitutively with both c-Cbl and CSF-1R. In addition, we show that expression of a dominant negative form of SLAP-2 impairs c-Cbl association with the CSF-1R and receptor ubiquitination. Impaired c-Cbl recruitment also correlated with changes in the kinetics of CSF-1R down-regulation and trafficking. CSF-1-mediated differentiation of FD-Fms cells and activation of downstream signaling events was also enhanced in cells stably expressing dominant negative SLAP-2. Together, these results demonstrate that SLAP-2 plays a role in c-Cbl-dependent down-regulation of CSF-1R signaling.
Collapse
Affiliation(s)
- Benjamin Pakuts
- Department of Medical Biophysics, University of Toronto, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Christophe Debonneville
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Larissa M Liontos
- Department of Medical Biophysics, University of Toronto, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Michael P Loreto
- Department of Medical Biophysics, University of Toronto, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - C Jane McGlade
- Department of Medical Biophysics, University of Toronto, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada; The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada.
| |
Collapse
|
2620
|
Alnaeeli M, Park J, Mahamed D, Penninger JM, Teng YTA. Dendritic cells at the osteo-immune interface: implications for inflammation-induced bone loss. J Bone Miner Res 2007; 22:775-80. [PMID: 17352656 DOI: 10.1359/jbmr.070314] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Within the past decade, the critical roles of T cells and T cell-mediated immunity in inflammation-induced osteoclastogenesis and subsequent bone loss have been extensively studied, thereby establishing the new paradigm of osteoimmunology. Therefore, dendritic cells (DCs), the most potent antigen-presenting cells, responsible for activation of naïve T cells and orchestration of the immune response, became critically situated at the osteo-immune interface. Today, emerging new evidence suggests that DC may be directly involved in inflammation-induced osteoclastogenesis and bone loss, by acting as osteoclast (OC) precursors that can further develop into DC-derived OCs (DDOC) under inflammatory conditions. These findings have tremendous implications, because in addition to DC's important roles in regulating innate and adaptive immunity, a direct contribution by these cells to inflammation-induced bone loss may provide a promising therapeutic target not only for controlling inflammation but also for modulating bone destruction.
Collapse
Affiliation(s)
- Mawadda Alnaeeli
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14620, USA
| | | | | | | | | |
Collapse
|
2621
|
Roberts HC, Knott L, Avery NC, Cox TM, Evans MJ, Hayman AR. Altered collagen in tartrate-resistant acid phosphatase (TRAP)-deficient mice: a role for TRAP in bone collagen metabolism. Calcif Tissue Int 2007; 80:400-10. [PMID: 17551769 DOI: 10.1007/s00223-007-9032-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 04/02/2007] [Accepted: 04/04/2007] [Indexed: 10/23/2022]
Abstract
Tartrate-resistant acid phosphatase (TRAP) is an iron-containing protein that is highly expressed by osteoclasts, macrophages, and dendritic cells. The enzyme is secreted by osteoclasts during bone resorption, and serum TRAP activity correlates with resorptive activity in disorders of bone metabolism. TRAP is essential for normal skeletal development. In knockout mice lacking TRAP, bone shape and modeling is altered with increased mineral density. Here, we report the effect of TRAP on the biochemical and biomechanical properties of collagen, the major protein constituting the bone matrix, using these mice. Femurs from TRAP-/- and wild-type mice were used in these studies. The biomechanical properties were investigated using a three-point bending technique. Collagen synthesis was determined by measuring cross-link content using high-performance liquid chromatography and amino acid analysis. Collagen degradation was determined by measuring matrix metalloproteinase-2 (MMP-2) activity. The rates of collagen synthesis and degradation were significantly greater in bones from TRAP-/- mice compared with wild type. At 8 weeks, there was an increase in the intermediate cross-links but no significant difference in animals aged 6 months. There was a significant increase in mature cross-links at both ages. A significant increase in MMP-2 production both pro and active was observed. A significant increase in ultimate stress and Young's modulus of elasticity was needed to fracture the bones from mice deficient in TRAP. We conclude that both synthesis as well as degradation of collagen are increased when TRAP is absent in mice at 8 weeks and 6 months of age, showing that TRAP has an important role in the metabolism of collagen.
Collapse
Affiliation(s)
- Helen C Roberts
- School of Clinical Veterinary Science, University of Bristol, Langford, BS40 5DU, UK
| | | | | | | | | | | |
Collapse
|
2622
|
Rajaraman G, Murthi P, Leo B, Brennecke SP, Kalionis B. Homeobox gene HLX1 is a regulator of colony stimulating factor-1 dependent trophoblast cell proliferation. Placenta 2007; 28:991-8. [PMID: 17532041 DOI: 10.1016/j.placenta.2007.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 03/23/2007] [Accepted: 03/30/2007] [Indexed: 11/18/2022]
Abstract
The cytokine colony stimulating factor-1 (CSF-1) is a key regulator of the proliferation, differentiation and activation of mononuclear phagocytes. CSF-1 also plays an important role in reproduction. CSF-1 is produced in the placenta and activates signal transduction pathways that significantly increase the proliferation of placental trophoblast cells in culture. The target genes activated by CSF-1 mediated signal transduction in the nucleus are not well understood. Here, we use placental trophoblast cells to investigate potential downstream effector genes of CSF-1. HLX1 is a homeobox gene that controls proliferation in embryonic cell types and haematopoietic cell lineages. We have shown HLX1 is expressed in placental trophoblast cells but its functional role in the placenta is unknown. Following CSF-1 stimulation, HLX1 mRNA expression was significantly increased in SGHPL-4 and HTR-8/SVNeo cultured trophoblast cells (p<0.001, n=3). siRNA-mediated reduction of HLX1 mRNA levels with four independent oligonucleotides (siRNAs) resulted in significantly decreased cell proliferation in both cell lines (p<0.001, n=4). When HLX1 mRNA levels were reduced in the presence of CSF-1 stimulation, proliferation remained significantly decreased (p<0.001, n=4) in both the cell lines. We have shown for the first time that a homeobox gene, HLX1, is a downstream effector gene of CSF-1, that HLX1 regulates placental cell proliferation and that CSF-1 acts, at least in part, through HLX1 to control cell proliferation.
Collapse
Affiliation(s)
- G Rajaraman
- Department of Obstetrics and Gynaecology, University of Melbourne, Australia
| | | | | | | | | |
Collapse
|
2623
|
Robays LJ, Maes T, Lebecque S, Lira SA, Kuziel WA, Brusselle GG, Joos GF, Vermaelen KV. Chemokine receptor CCR2 but not CCR5 or CCR6 mediates the increase in pulmonary dendritic cells during allergic airway inflammation. THE JOURNAL OF IMMUNOLOGY 2007; 178:5305-11. [PMID: 17404315 DOI: 10.4049/jimmunol.178.8.5305] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increased numbers of pulmonary dendritic cells (DCs) are recruited to the lungs during allergic airway inflammation and contribute to the maintenance of the inflammatory immune response. The chemokine receptors that directly control DC accumulation into the lungs are largely unknown. To explore this issue, we generated mixed bone marrow chimeric mice containing both wild-type and knockout cells for a given chemokine receptor. After induction of allergic airway inflammation, we specifically tracked and compared chemokine receptor knockout vs wild-type DC populations through various lung compartments. Using this approach, we show that CCR2, but not CCR5 or CCR6, directly controls the accumulation of DCs into allergic lungs. Furthermore, the size of inflammatory monocyte populations in peripheral blood was strikingly CCR2 dependent, suggesting that CCR2 primarily mediates the release of monocytic DC precursors into the bloodstream.
Collapse
Affiliation(s)
- Lander J Robays
- Department of Respiratory Diseases, Ghent University Hospital, Ghent, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
2624
|
Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. ACTA ACUST UNITED AC 2007; 204:1057-69. [PMID: 17485518 PMCID: PMC2118577 DOI: 10.1084/jem.20070075] [Citation(s) in RCA: 1528] [Impact Index Per Article: 84.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages (MPs) are important for skeletal muscle regeneration in vivo and may exert beneficial effects on myogenic cell growth through mitogenic and antiapoptotic activities in vitro. However, MPs are highly versatile and may exert various, and even opposite, functions depending on their activation state. We studied monocyte (MO)/MP phenotypes and functions during skeletal muscle repair. Selective labeling of circulating MOs by latex beads in CX3CR1(GFP/+) mice showed that injured muscle recruited only CX3CR1(lo)/Ly-6C(+) MOs from blood that exhibited a nondividing, F4/80(lo), proinflammatory profile. Then, within muscle, these cells switched their phenotype to become proliferating antiinflammatory CX3CR1(hi)/Ly-6C(-) cells that further differentiated into F4/80(hi) MPs. In vitro, phagocytosis of muscle cell debris induced a switch of proinflammatory MPs toward an antiinflammatory phenotype releasing transforming growth factor beta1. In co-cultures, inflammatory MPs stimulated myogenic cell proliferation, whereas antiinflammatory MPs exhibited differentiating activity, assessed by both myogenin expression and fusion into myotubes. Finally, depletion of circulating MOs in CD11b-diphtheria toxin receptor mice at the time of injury totally prevented muscle regeneration, whereas depletion of intramuscular F4/80(hi) MPs at later stages reduced the diameter of regenerating fibers. In conclusion, injured skeletal muscle recruits MOs exhibiting inflammatory profiles that operate phagocytosis and rapidly convert to antiinflammatory MPs that stimulate myogenesis and fiber growth.
Collapse
Affiliation(s)
- Ludovic Arnold
- Institut National de la Santé et de la Recherche Médicale, Unité 841, Institut Mondor de Recherche Biomédicale, Cell Interactions in the Neuromuscular System Team, Université Paris 12 Val-de-Marne, Créteil, France
| | | | | | | | | | | | | | | |
Collapse
|
2625
|
Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol 2007; 82:244-52. [PMID: 17475785 DOI: 10.1189/jlb.0307191] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The term "monocyte" implies a single, homogenous population of cells with uniform physiology. Recent evidence from a number of laboratories indicates that it is likely that blood monocytes may consist of several subpopulations of cells, which differ in size, nuclear morphology, granularity, and functionality. The aim of this review is to give a summary of the new findings in the emerging field of monocyte heterogeneity. We provide a short description of the differentiation patterns of blood monocyte subpopulations, with an emphasis on how these subpopulations can be influenced by infection. We provide a comparison among the main monocyte subpopulations in humans, mice, and rats and illustrate some of the common features of these cells and some of the important interspecies distinctions. We will also discuss the bone marrow precursors of these cells and the differentiation patterns of these subsets in different tissues in response to infection. Most of the data about monocyte trafficking during infection are necessarily derived from murine models, and comparisons between mouse and man must be made with caution. However, these models may provide interesting springboards to permit us to speculate about the topic of monocyte heterogeneity in humans.
Collapse
Affiliation(s)
- Dalit Strauss-Ayali
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, USA
| | | | | |
Collapse
|
2626
|
Markey MP, Bergseid J, Bosco EE, Stengel K, Xu H, Mayhew CN, Schwemberger SJ, Braden WA, Jiang Y, Babcock GF, Jegga AG, Aronow BJ, Reed MF, Wang JYJ, Knudsen ES. Loss of the retinoblastoma tumor suppressor: differential action on transcriptional programs related to cell cycle control and immune function. Oncogene 2007; 26:6307-18. [PMID: 17452985 DOI: 10.1038/sj.onc.1210450] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Functional inactivation of the retinoblastoma tumor suppressor gene product (RB) is a common event in human cancers. Classically, RB functions to constrain cellular proliferation, and loss of RB is proposed to facilitate the hyperplastic proliferation associated with tumorigenesis. To understand the repertoire of regulatory processes governed by RB, two models of RB loss were utilized to perform microarray analysis. In murine embryonic fibroblasts harboring germline loss of RB, there was a striking deregulation of gene expression, wherein distinct biological pathways were altered. Specifically, genes involved in cell cycle control and classically associated with E2F-dependent gene regulation were upregulated via RB loss. In contrast, a program of gene expression associated with immune function and response to pathogens was significantly downregulated with the loss of RB. To determine the specific influence of RB loss during a defined period and without the possibility of developmental compensation as occurs in embryonic fibroblasts, a second system was employed wherein Rb was acutely knocked out in adult fibroblasts. This model confirmed the distinct regulation of cell cycle and immune modulatory genes through RB loss. Analyses of cis-elements supported the hypothesis that the majority of those genes upregulated with RB loss are regulated via the E2F family of transcription factors. In contrast, those genes whose expression was reduced with the loss of RB harbored different promoter elements. Consistent with these analyses, we found that disruption of E2F-binding function of RB was associated with the upregulation of gene expression. In contrast, cells harboring an RB mutant protein (RB-750F) that retains E2F-binding activity, but is specifically deficient in the association with LXCXE-containing proteins, failed to upregulate these same target genes. However, downregulation of genes involved in immune function was readily observed with disruption of the LXCXE-binding function of RB. Thus, these studies demonstrate that RB plays a significant role in both the positive and negative regulations of transcriptional programs and indicate that loss of RB has distinct biological effects related to both cell cycle control and immune function.
Collapse
Affiliation(s)
- M P Markey
- Department of Cell and Cancer Biology, University of Cincinnati, Cincinnati, OH 45267-0521, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2627
|
Boos MD, Yokota Y, Eberl G, Kee BL. Mature natural killer cell and lymphoid tissue-inducing cell development requires Id2-mediated suppression of E protein activity. ACTA ACUST UNITED AC 2007; 204:1119-30. [PMID: 17452521 PMCID: PMC2118569 DOI: 10.1084/jem.20061959] [Citation(s) in RCA: 301] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Id2 transcriptional repressor is essential for development of natural killer (NK) cells, lymphoid tissue–inducing (LTi) cells, and secondary lymphoid tissues. Id2 was proposed to regulate NK and LTi lineage specification from multipotent progenitors through suppression of E proteins. We report that NK cell progenitors are not reduced in the bone marrow (BM) of Id2−/− mice, demonstrating that Id2 is not essential for NK lineage specification. Rather, Id2 is required for development of mature (m) NK cells. We define the mechanism by which Id2 functions by showing that a reduction in E protein activity, through deletion of E2A, overcomes the need for Id2 in development of BM mNK cells, LTi cells, and secondary lymphoid tissues. However, mNK cells are not restored in the blood or spleen of Id2−/−E2A−/− mice, suggesting a role for Id2 in suppression of alternative E proteins after maturation. Interestingly, the few splenic mNK cells in Id2−/− and Id2−/−E2A−/− mice have characteristics of thymus-derived NK cells, which develop in the absence of Id2, implying a differential requirement for Id2 in BM and thymic mNK development. Our findings redefine the essential functions of Id2 in lymphoid development and provide insight into the dynamic regulation of E and Id proteins during this process.
Collapse
Affiliation(s)
- Markus D Boos
- Committee on Immunology, University of Chicago, Chicago, IL 60657, USA
| | | | | | | |
Collapse
|
2628
|
Liu K, Waskow C, Liu X, Yao K, Hoh J, Nussenzweig M. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat Immunol 2007; 8:578-83. [PMID: 17450143 DOI: 10.1038/ni1462] [Citation(s) in RCA: 352] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Accepted: 04/03/2007] [Indexed: 12/13/2022]
Abstract
Parabiosis experiments demonstrating that dendritic cells (DCs) do not equilibrate between mice even after prolonged joining by parabiosis have suggested that DCs are derived from self-renewing progenitors that divide in situ. However, here we found that unequal exchange of DCs between mice joined by parabiosis reflected uneven distribution of DC precursors in blood due to their short half-life in circulation. DCs underwent only a limited number of divisions in the spleen or lymph nodes over a 10- to 14-day period and were replenished from blood-borne precursors at a rate of nearly 4,300 cells per hour. Daughter DCs presented antigens captured by their progenitors, suggesting that DC division in peripheral lymphoid organs can prolong the duration of antigen presentation in vivo.
Collapse
Affiliation(s)
- Kang Liu
- Laboratory of Molecular Immunology, The Rockefeller University, New York, New York 10021, USA
| | | | | | | | | | | |
Collapse
|
2629
|
Liu XS, Li XH, Wang Y, Shu RZ, Wang L, Lu SY, Kong H, Jin YE, Zhang LJ, Fei J, Chen SJ, Chen Z, Gu MM, Lu ZY, Wang ZG. Disruption of palladin leads to defects in definitive erythropoiesis by interfering with erythroblastic island formation in mouse fetal liver. Blood 2007; 110:870-6. [PMID: 17431131 DOI: 10.1182/blood-2007-01-068528] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Palladin was originally found up-regulated with NB4 cell differentiation induced by all-trans retinoic acid. Disruption of palladin results in neural tube closure defects, liver herniation, and embryonic lethality. Here we further report that Palld(-/-) embryos exhibit a significant defect in erythropoiesis characterized by a dramatic reduction in definitive erythrocytes derived from fetal liver but not primitive erythrocytes from yolk sac. The reduction of erythrocytes is accompanied by increased apoptosis of erythroblasts and partial blockage of erythroid differentiation. However, colony-forming assay shows no differences between wild-type (wt) and mutant fetal liver or yolk sac in the number and size of colonies tested. In addition, Palld(-/-) fetal liver cells can reconstitute hematopoiesis in lethally irradiated mice. These data strongly suggest that deficient erythropoiesis in Palld(-/-) fetal liver is mainly due to a compromised erythropoietic microenvironment. As expected, erythroblastic island in Palld(-/-) fetal liver was found disorganized. Palld(-/-) fetal liver cells fail to form erythroblastic island in vitro. Interestingly, wt macrophages can form such units with either wt or mutant erythroblasts, while mutant macrophages lose their ability to bind wt or mutant erythroblasts. These data demonstrate that palladin is crucial for definitive erythropoiesis and erythroblastic island formation and, especially, required for normal function of macrophages in fetal liver.
Collapse
Affiliation(s)
- Xue-Song Liu
- Laboratory of Genetic Engineering, Department of Medical Genetics, Institute of Health Sciences, Shanghai Institutes for Biological Sciences/Chinese Academy of Sciences, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2630
|
León B, López-Bravo M, Ardavín C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 2007; 26:519-31. [PMID: 17412618 DOI: 10.1016/j.immuni.2007.01.017] [Citation(s) in RCA: 522] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 01/22/2007] [Accepted: 01/29/2007] [Indexed: 01/24/2023]
Abstract
Infection-induced inflammatory reactions involve a strong increase in dendritic cells (DCs) at the infection site and draining lymph nodes (dLNs). Whether inflammatory DCs are recruited to these locations or differentiate locally, and what their functional relevance is, remain unclear. Here we showed that during Leishmania infection, monocytes were recruited to the dermis and differentiated into "dermal monocyte-derived DCs," which subsequently migrated into the dLNs. In addition, monocyte recruitment to the dLNs resulted in the differentiation into "LN monocyte-derived DCs." Analysis of the kinetics of monocyte differentiation into DCs, susceptibility to infection, IL-12 production, and L. major-specific T cell stimulation potential suggest that dermal monocyte-derived DCs controlled the induction of protective T helper 1 responses against Leishmania. Thus, the demonstration of monocyte differentiation potential into DCs during in vivo infection and of local DC differentiation in inflammatory foci suggests that de novo formed monocyte-derived DCs are essential in T cell immunity against pathogens.
Collapse
Affiliation(s)
- Beatriz León
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Universidad Autónoma, 28049 Madrid, Spain
| | | | | |
Collapse
|
2631
|
Abstract
The combination of the induction of lymphopenia and vaccination and/or T cell transfer is garnering much attention for cancer treatment. Preclinical studies have shown that the induction of lymphopenia by chemotherapy or radiation can enhance the antitumor efficacy of several distinct, cell-based immunotherapeutic approaches. The mechanism(s) by which such enhancement is achieved are being intensively studied, yet there is much opportunity for improvement. The animal studies reported by Wrzesinski and colleagues in this issue of the JCI are a promising and timely step in this direction (see the related article beginning on page 492). The authors have evaluated both the effect of increasing the intensity of lymphodepletion and the influence of HSC transfer on the in vivo function of adoptively transferred CD8(+) T cells. We discuss their results in light of the evolving field and their implications for advancing cell-based immunotherapies for cancer.
Collapse
Affiliation(s)
- Claudio Anasetti
- H. Lee Moffitt Comprehensive Cancer Center, Tampa, Florida 33612, USA
| | | |
Collapse
|
2632
|
Karsdal MA, Martin TJ, Bollerslev J, Christiansen C, Henriksen K. Are nonresorbing osteoclasts sources of bone anabolic activity? J Bone Miner Res 2007; 22:487-94. [PMID: 17227224 DOI: 10.1359/jbmr.070109] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Some osteopetrotic mutations lead to low resorption, increased numbers of osteoclasts, and increased bone formation, whereas other osteopetrotic mutations lead to low resorption, low numbers of osteoclasts, and decreased bone formation. Elaborating on these findings, we discuss the possibility that osteoclasts are the source of anabolic signals for osteoblasts. In normal healthy individuals, bone formation is coupled to bone resorption in a tight equilibrium. When this delicate balance is disturbed, the net result is pathological situations, such as osteopetrosis or osteoporosis. Human osteopetrosis, caused by mutations in proteins involved in the acidification of the resorption lacuna (ClC-7 or the a3-V-ATPase), is characterized by decreased resorption in face of normal or even increased bone formation. Mouse mutations leading to ablation of osteoclasts (e.g., loss of macrophage-colony stimulating factor [M-CSF] or c-fos) lead to secondary negative effects on bone formation, in contrast to mutations where bone resorption is abrogated with sustained osteoclast numbers, such as the c-src mice. These data indicate a central role for osteoclasts, and not necessarily their resorptive activity, in the control of bone formation. In this review, we consider the balance between bone resorption and bone formation, reviewing novel data that have shown that this principle is more complex than originally thought. We highlight the distinct possibility that osteoclast function can be divided into two more or less separate functions, namely bone resorption and stimulation of bone formation. Finally, we describe the likely possibility that bone resorption can be attenuated pharmacologically without the undesirable reduction in bone formation.
Collapse
|
2633
|
Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, Lira SA, Habenicht AJ, Randolph GJ. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest 2007; 117:185-94. [PMID: 17200718 PMCID: PMC1716202 DOI: 10.1172/jci28549] [Citation(s) in RCA: 1074] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 10/24/2006] [Indexed: 12/12/2022] Open
Abstract
Monocytes participate critically in atherosclerosis. There are 2 major subsets expressing different chemokine receptor patterns: CCR2(+)CX3CR1(+)Ly-6C(hi) and CCR2(-)CX3CR1(++)Ly-6C(lo) monocytes. Both C-C motif chemokine receptor 2 (CCR2) and C-X(3)-C motif chemokine receptor 1 (CX3CR1) are linked to progression of atherosclerotic plaques. Here, we analyzed mouse monocyte subsets in apoE-deficient mice and traced their differentiation and chemokine receptor usage as they accumulated within atherosclerotic plaques. Blood monocyte counts were elevated in apoE(-/-) mice and skewed toward an increased frequency of CCR2(+)Ly-6C(hi) monocytes in apoE(-/-) mice fed a high-fat diet. CCR2(+)Ly-6C(hi) monocytes efficiently accumulated in plaques, whereas CCR2(-)Ly-6C(lo) monocytes entered less frequently but were more prone to developing into plaque cells expressing the dendritic cell-associated marker CD11c, indicating that phagocyte heterogeneity in plaques is linked to distinct types of entering monocytes. CCR2(-) monocytes did not rely on CX3CR1 to enter plaques. Instead, they were partially dependent upon CCR5, which they selectively upregulated in apoE(-/-) mice. By comparison, CCR2(+)Ly-6C(hi) monocytes unexpectedly required CX3CR1 in addition to CCR2 and CCR5 to accumulate within plaques. In many other inflammatory settings, these monocytes utilize CCR2, but not CX3CR1, for trafficking. Thus, antagonizing CX3CR1 may be effective therapeutically in ameliorating CCR2(+) monocyte recruitment to plaques without impairing their CCR2-dependent responses to inflammation overall.
Collapse
Affiliation(s)
- Frank Tacke
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - David Alvarez
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Theodore J. Kaplan
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Claudia Jakubzick
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Rainer Spanbroek
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Jaime Llodra
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Alexandre Garin
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Jianhua Liu
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Matthias Mack
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Nico van Rooijen
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Sergio A. Lira
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Andreas J. Habenicht
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - Gwendalyn J. Randolph
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York, USA.
Institute for Vascular Medicine, Friedrich-Schiller-University, Jena, Germany.
Department of Immunobiology, Icahn Medical Institute, and
The Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, New York, USA.
Department of Internal Medicine, University Clinic, University of Regensburg, Regensburg, Germany.
Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
2634
|
Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, Pittet MJ. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest 2007; 117:195-205. [PMID: 17200719 PMCID: PMC1716211 DOI: 10.1172/jci29950] [Citation(s) in RCA: 1004] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 10/24/2006] [Indexed: 12/14/2022] Open
Abstract
Macrophage accumulation participates decisively in the development and exacerbation of atherosclerosis. Circulating monocytes, the precursors of macrophages, display heterogeneity in mice and humans, but their relative contribution to atherogenesis remains unknown. We report here that the Ly-6C(hi) monocyte subset increased dramatically in hypercholesterolemic apoE-deficient mice consuming a high-fat diet, with the number of Ly-6C(hi) cells doubling in the blood every month. Ly-6C(hi) monocytes adhered to activated endothelium, infiltrated lesions, and became lesional macrophages. Hypercholesterolemia-associated monocytosis (HAM) developed from increased survival, continued cell proliferation, and impaired Ly-6C(hi) to Ly-6C(lo) conversion and subsided upon statin-induced cholesterol reduction. Conversely, the number of Ly-6C(lo) cells remained unaffected. Thus, we believe that Ly-6C(hi) monocytes represent a newly recognized component of the inflammatory response in experimental atherosclerosis.
Collapse
Affiliation(s)
- Filip K Swirski
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | | | | | | | | | | | |
Collapse
|
2635
|
Jori FP, Galderisi U, Napolitano MA, Cipollaro M, Cascino A, Giordano A, Melone MAB. RB and RB2/P130 genes cooperate with extrinsic signals to promote differentiation of rat neural stem cells. Mol Cell Neurosci 2007; 34:299-309. [PMID: 17223573 DOI: 10.1016/j.mcn.2006.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 10/04/2006] [Accepted: 11/13/2006] [Indexed: 11/20/2022] Open
Abstract
Mechanisms governing commitment and differentiation of the cells of the nervous system begin to be elucidated: how extrinsic and intrinsic components are related remains poorly understood. To investigate this issue, we overexpressed genes of the retinoblastoma (Rb) family RB and RB2/p130, which play an important role during nerve cell maturation, in rat neural stem cells (NSCs). Immunostaining of neurons, astrocytes and oligodendrocytes in cultures overexpressing pRb or pRb2/p130 revealed that these genes affect lineage specification of differentiating NSCs. We observed modifications in percentage of differentiated cells indicating a shift towards the phenotype induced by culture conditions. Results were confirmed by detection of the expression levels of differentiation markers by RT-PCR. Analysis of BrdU incorporation and detection of an early marker of apoptosis suggest that the effect of pRb and pRb2/p130 overexpression is not dependent on the inhibition of cell proliferation, nor does it rely on the regulation of cell survival. Our findings suggest that Rb family genes are involved in fate determination of the cells of the nervous system. However, their role seems subsidiary to that of the extrinsic signals that promote lineage specification and appear to be mediated by a direct effect on the acquisition of a specific phenotype.
Collapse
Affiliation(s)
- Francesco P Jori
- Department of Neurological Sciences, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
2636
|
Ohno H, Kubo K, Murooka H, Kobayashi Y, Nishitoba T, Shibuya M, Yoneda T, Isoe T. A c-fms tyrosine kinase inhibitor, Ki20227, suppresses osteoclast differentiation and osteolytic bone destruction in a bone metastasis model. Mol Cancer Ther 2007; 5:2634-43. [PMID: 17121910 DOI: 10.1158/1535-7163.mct-05-0313] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In bone metastatic lesions, osteoclasts play a key role in the development of osteolysis. Previous studies have shown that macrophage colony-stimulating factor (M-CSF) is important for the differentiation of osteoclasts. In this study, we investigated whether an inhibitor of M-CSF receptor (c-Fms) suppresses osteoclast-dependent osteolysis in bone metastatic lesions. We developed small molecule inhibitors against ligand-dependent phosphorylation of c-Fms and examined the effects of these compounds on osteolytic bone destruction in a bone metastasis model. We discovered a novel quinoline-urea derivative, Ki20227 (N-{4-[(6,7-dimethoxy-4-quinolyl)oxy]-2-methoxyphenyl}-N'-[1-(1,3-thiazole-2-yl)ethyl]urea), which is a c-Fms tyrosine kinase inhibitor. The IC(50)s of Ki20227 to inhibit c-Fms, vascular endothelial growth factor receptor-2 (KDR), stem cell factor receptor (c-Kit), and platelet-derived growth factor receptor beta were found to be 2, 12, 451, and 217 nmol/L, respectively. Ki20227 did not inhibit other kinases tested, such as fms-like tyrosine kinase-3, epidermal growth factor receptor, or c-Src (c-src proto-oncogene product). Ki20227 was also found to inhibit the M-CSF-dependent growth of M-NFS-60 cells but not the M-CSF-independent growth of A375 human melanoma cells in vitro. Furthermore, in an osteoclast-like cell formation assay using mouse bone marrow cells, Ki20227 inhibited the development of tartrate-resistant acid phosphatase-positive osteoclast-like cells in a dose-dependent manner. In in vivo studies, oral administration of Ki20227 suppressed osteoclast-like cell accumulation and bone resorption induced by metastatic tumor cells in nude rats following intracardiac injection of A375 cells. Moreover, Ki20227 decreased the number of tartrate-resistant acid phosphatase-positive osteoclast-like cells on bone surfaces in ovariectomized (ovx) rats. These findings suggest that Ki20227 inhibits osteolytic bone destruction through the suppression of M-CSF-induced osteoclast accumulation in vivo. Therefore, Ki20227 may be a useful therapeutic agent for osteolytic disease associated with bone metastasis and other bone diseases.
Collapse
Affiliation(s)
- Hiroaki Ohno
- Pharmaceutical Research Laboratories, Kirin Brewery Co., Ltd., 3 Miyahara, Takasaki, Gunma, 370-1295, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
2637
|
Landsman L, Varol C, Jung S. Distinct Differentiation Potential of Blood Monocyte Subsets in the Lung. THE JOURNAL OF IMMUNOLOGY 2007; 178:2000-7. [PMID: 17277103 DOI: 10.4049/jimmunol.178.4.2000] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Peripheral blood monocytes are a population of circulating mononuclear phagocytes that harbor potential to differentiate into macrophages and dendritic cells. As in humans, monocytes in the mouse comprise two phenotypically distinct subsets that are Gr1(high)CX(3)CR1(int) and Gr1(low)CX(3)CR1(high), respectively. The question remains whether these populations contribute differentially to the generation of peripheral mononuclear phagocytes. In this study, we track the fate of adoptively transferred, fractionated monocyte subsets in the lung of recipient mice. We show that under inflammatory and noninflammatory conditions, both monocyte subsets give rise to pulmonary dendritic cells. In contrast, under the conditions studied, only Gr1(low)CX(3)CR1(high) monocytes, but not Gr1(high)CX(3)CR1(int) cells, had the potential to differentiate into lung macrophages. However, Gr1(high)CX(3)CR1(int) monocytes could acquire this potential upon conversion into Gr1(low)CX(3)CR1(high) cells. Our results therefore indicate an intrinsic dichotomy in the differentiation potential of the two main blood monocyte subsets.
Collapse
Affiliation(s)
- Limor Landsman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
2638
|
Karsdal MA, Qvist P, Christiansen C, Tankó LB. Optimising antiresorptive therapies in postmenopausal women: why do we need to give due consideration to the degree of suppression? Drugs 2007; 66:1909-18. [PMID: 17100403 DOI: 10.2165/00003495-200666150-00002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Accelerated bone turnover with bone resorption exceeding bone formation is a major mechanism underlying postmenopausal bone loss and hence the development of osteoporosis. Accordingly, inhibition of bone resorption is a rational approach for the prevention of osteoporosis. In this context, the most logical option, hormone replacement therapy, reverses the rate of bone turnover to premenopausal levels, whereas the magnitude of inhibition by amino-bisphosphonates and the recently introduced anti-receptor activator of NFkappaB ligand (RANKL) antibody often exceeds this. As bone turnover has crucial implications for the continuous renewal of bone tissue, the over-suppression of bone turnover has potential consequences for bone quality and strength. Long-term treatment with potent bisphosphonates has recently been associated with osteonecrosis of the jaw and dose-dependent increases in micro-crack accumulation in animals. Although these observations are the subject of ongoing discussions, it is timely to discuss whether the over-suppression of bone turnover below premenopausal levels is really our ultimate goal when defining the success criteria for antiresorptive agents. In this review, the implications of high and excessively low bone turnover of endogenous origin for bone quality, fracture risk and integrity of the jaw are discussed. In addition, animal and clinical research revealing initial findings regarding the potential adverse effects of drug-induced suppression of bone remodeling are summarised. The inhibition of bone resorption, which is either transient between doses (e.g. with calcitonin) or does not exceed premenopausal levels (with hormone replacement therapy or selective estrogen receptor modulators), is preferable because it not only provides similar antifracture efficacy but can also assist in the maintenance of the dynamic repair of micro-cracks/micro-fractures.
Collapse
|
2639
|
Asagiri M, Takayanagi H. The molecular understanding of osteoclast differentiation. Bone 2007; 40:251-64. [PMID: 17098490 DOI: 10.1016/j.bone.2006.09.023] [Citation(s) in RCA: 1057] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 09/01/2006] [Accepted: 09/06/2006] [Indexed: 12/12/2022]
Abstract
Osteoclasts are multinucleated cells of monocyte/macrophage origin that degrade bone matrix. The differentiation of osteoclasts is dependent on a tumor necrosis factor (TNF) family cytokine, receptor activator of nuclear factor (NF)-kappaB ligand (RANKL), as well as macrophage colony-stimulating factor (M-CSF). Congenital lack of osteoclasts causes osteopetrosis, investigation of which has provided insights into the essential molecules for osteoclastogenesis, including TNF receptor-associated factor (TRAF) 6, NF-kappaB and c-Fos. In addition, genome-wide screening techniques have shed light on an additional set of gene products such as nuclear factor of activated T cells (NFAT) c1. Here we summarize the efforts to understand the sequential molecular events induced by RANKL during osteoclast differentiation. RANKL binds to its receptor RANK, which recruits adaptor molecules such as TRAF6. TRAF6 activates NF-kappaB, which is important for the initial induction of NFATc1. NFATc1 is activated by calcium signaling and binds to its own promoter, thus switching on an autoregulatory loop. An activator protein (AP)-1 complex containing c-Fos is required for the autoamplification of NFATc1, enabling the robust induction of NFATc1. Finally, NFATc1 cooperates with other transcriptional partners to activate osteoclast-specific genes. NFATc1 autoregulation is controlled by an epigenetic mechanism, which has profound implications for an understanding of the general mechanism of irreversible cell fate determination. From the clinical point of view, RANKL signaling pathway has promise as a strategy for suppressing the excessive osteoclast formation characteristic of a variety of bone diseases.
Collapse
Affiliation(s)
- Masataka Asagiri
- Department of Cell Signaling, Graduate School, Tokyo Medical and Dental University, Yushima 1-5-45, Tokyo 113-8549, Japan
| | | |
Collapse
|
2640
|
Marshall D, Cameron J, Lightwood D, Lawson ADG. Blockade of colony stimulating factor-1 (CSF-I) leads to inhibition of DSS-induced colitis. Inflamm Bowel Dis 2007; 13:219-24. [PMID: 17206685 DOI: 10.1002/ibd.20055] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal inflammation associated with inflammatory bowel disease (IBD) is typically characterized by an inflammatory cell infiltrate and pro-inflammatory cytokine production. Of particular interest, the frequency of colony stimulating factor-1 (CSF-l)-expressing cells is increased in active lesions. In this study, we have investigated the role of CSF-1 in mucosal inflammation, using a murine model of colitis induced by dextran sulfate sodium (DSS). METHODS A neutralizing anti-CSF-1 antibody was administered to Balb/c mice that received DSS in their drinking water. Signs of colitis, such as clinical disease score, cellular infiltrate, and cytokine production, were assessed. RESULTS Administration of a neutralizing anti-CSF-1 antibody significantly inhibited DSS-induced colitis. Clinical symptoms, such as weight loss and the appearance of diarrhea or fecal blood, were reduced by CSF-1 blockade; histologic scores were also improved. The cellular infiltrate of macrophages and T cells was inhibited and a trend toward reduced production of pro-inflammatory cytokines was noted. CONCLUSIONS This is the first study to demonstrate that CSF-1 plays an important role in mediating intestinal mucosal inflammation and therefore may prove to be an attractive therapeutic target for intestinal diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Diane Marshall
- Celltech Centre of Excellence for Antibody Research, UCB, 216 Bath Road, Slough SLI 4EN, UK.
| | | | | | | |
Collapse
|
2641
|
Schubert C, Schalk-Hihi C, Struble GT, Ma HC, Petrounia IP, Brandt B, Deckman IC, Patch RJ, Player MR, Spurlino JC, Springer BA. Crystal Structure of the Tyrosine Kinase Domain of Colony-stimulating Factor-1 Receptor (cFMS) in Complex with Two Inhibitors. J Biol Chem 2007; 282:4094-101. [PMID: 17132624 DOI: 10.1074/jbc.m608183200] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cFMS proto-oncogene encodes for the colony-stimulating factor-1 receptor, a receptor-tyrosine kinase responsible for the differentiation and maturation of certain macrophages. Upon binding its ligand colony-stimulating factor-1 cFMS autophosphorylates, dimerizes, and induces phosphorylation of downstream targets. We report the novel crystal structure of unphosphorylated cFMS in complex with two members of different classes of drug-like protein kinase inhibitors. cFMS exhibits a typical bi-lobal kinase fold, and its activation loop and DFG motif are found to be in the canonical inactive conformation. Both ATP competitive inhibitors are bound in the active site and demonstrate a binding mode similar to that of STI-571 bound to cABL. The DFG motif is prevented from switching into the catalytically competent conformation through interactions with the inhibitors. Activation of cFMS is also inhibited by the juxtamembrane domain, which interacts with residues of the active site and prevents formation of the activated kinase. Together the structures of cFMS provide further insight into the autoinhibition of receptor-tyrosine kinases via their respective juxtamembrane domains; additionally the binding mode of two novel classes of kinase inhibitors will guide the design of novel molecules targeting macrophage-related diseases.
Collapse
MESH Headings
- Amides/chemistry
- Amino Acid Sequence
- Binding Sites
- Crystallography, X-Ray
- Humans
- Molecular Sequence Data
- Mutant Chimeric Proteins/antagonists & inhibitors
- Mutant Chimeric Proteins/chemistry
- Protein Kinase Inhibitors/chemistry
- Protein Structure, Tertiary/genetics
- Proto-Oncogene Mas
- Quinolones/chemistry
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/chemistry
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptor, Macrophage Colony-Stimulating Factor/chemistry
- Receptor, Macrophage Colony-Stimulating Factor/genetics
- Receptor, Macrophage Colony-Stimulating Factor/metabolism
- Receptor, TIE-2/chemistry
- Receptor, TIE-2/genetics
- Receptors, Fibroblast Growth Factor/chemistry
- Receptors, Fibroblast Growth Factor/genetics
Collapse
Affiliation(s)
- Carsten Schubert
- Structural Biology, Johnson & Johnson Pharmaceuticals Research and Development, LLC, Exton, Pennsylvania 19341, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2642
|
Wenzel PL, Wu L, de Bruin A, Chong JL, Chen WY, Dureska G, Sites E, Pan T, Sharma A, Huang K, Ridgway R, Mosaliganti K, Sharp R, Machiraju R, Saltz J, Yamamoto H, Cross JC, Robinson ML, Leone G. Rb is critical in a mammalian tissue stem cell population. Genes Dev 2007; 21:85-97. [PMID: 17210791 PMCID: PMC1759903 DOI: 10.1101/gad.1485307] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The inactivation of the retinoblastoma (Rb) tumor suppressor gene in mice results in ectopic proliferation, apoptosis, and impaired differentiation in extraembryonic, neural, and erythroid lineages, culminating in fetal death by embryonic day 15.5 (E15.5). Here we show that the specific loss of Rb in trophoblast stem (TS) cells, but not in trophoblast derivatives, leads to an overexpansion of trophoblasts, a disruption of placental architecture, and fetal death by E15.5. Despite profound placental abnormalities, fetal tissues appeared remarkably normal, suggesting that the full manifestation of fetal phenotypes requires the loss of Rb in both extraembryonic and fetal tissues. Loss of Rb resulted in an increase of E2f3 expression, and the combined ablation of Rb and E2f3 significantly suppressed Rb mutant phenotypes. This rescue appears to be cell autonomous since the inactivation of Rb and E2f3 in TS cells restored placental development and extended the life of embryos to E17.5. Taken together, these results demonstrate that loss of Rb in TS cells is the defining event causing lethality of Rb(-/-) embryos and reveal the convergence of extraembryonic and fetal functions of Rb in neural and erythroid development. We conclude that the Rb pathway plays a critical role in the maintenance of a mammalian stem cell population.
Collapse
Affiliation(s)
- Pamela L. Wenzel
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Lizhao Wu
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Alain de Bruin
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jean-Leon Chong
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Wen-Yi Chen
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Geoffrey Dureska
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Emily Sites
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Tony Pan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Ashish Sharma
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Kun Huang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Randall Ridgway
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Kishore Mosaliganti
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Richard Sharp
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Raghu Machiraju
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Joel Saltz
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Hideyuki Yamamoto
- Department of Biochemistry and Molecular Biology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - James C. Cross
- Department of Biochemistry and Molecular Biology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Michael L. Robinson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Division of Molecular and Human Genetics, Children’s Research Institute, Columbus, Ohio 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio 43210, USA
- E-MAIL ; FAX (513) 529-6900
| | - Gustavo Leone
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Corresponding authors.E-MAIL ; FAX (614) 292-3312
| |
Collapse
|
2643
|
Khanna-Gupta A, Sun H, Zibello T, Lee HM, Dahl R, Boxer LA, Berliner N. Growth factor independence-1 (Gfi-1) plays a role in mediating specific granule deficiency (SGD) in a patient lacking a gene-inactivating mutation in the C/EBPepsilon gene. Blood 2007; 109:4181-90. [PMID: 17244686 PMCID: PMC1885490 DOI: 10.1182/blood-2005-05-022004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neutrophil-specific granule deficiency (SGD) is a rare congenital disorder marked by recurrent bacterial infections. Neutrophils from SGD patients lack secondary and tertiary granules and their content proteins and lack normal neutrophil functions. Gene-inactivating mutations in the C/EBPepsilon gene have been identified in 2 SGD patients. Our studies on a third SGD patient revealed a heterozygous mutation in the C/EBPepsilon gene. However, we demonstrate elevated levels of C/EBPepsilon and PU.1 proteins in the patient's peripheral blood neutrophils. The expression of the transcription factor growth factor independence-1 (Gfi-1), however, was found to be markedly reduced in our SGD patient despite the absence of an obvious mutation in this gene. This may explain the elevated levels of both C/EBPepsilon and PU.1, which are targets of Gfi-1 transcriptional repression. We have generated a growth factor-dependent EML cell line from the bone marrow of Gfi-1(+/-) and Gfi-1(+/+) mice as a model for Gfi-1-deficient SGD, and demonstrate that lower levels of Gfi-1 expression in the Gfi-1(+/-) EML cells is associated with reduced levels of secondary granule protein (SGP) gene expression. Furthermore, we demonstrate a positive role for Gfi-1 in SGP expression, in that Gfi-1 binds to and up-regulates the promoter of neutrophil collagenase (an SGP gene), in cooperation with wild-type but not with mutant C/EBPepsilon. We hypothesize that decreased Gfi-1 levels in our SGD patient, together with the mutant C/EBPepsilon, block SGP expression, thereby contributing to the underlying etiology of the disease in our patient.
Collapse
Affiliation(s)
- Arati Khanna-Gupta
- Section of Hematology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | |
Collapse
|
2644
|
Furtado GC, Piña B, Tacke F, Gaupp S, van Rooijen N, Moran TM, Randolph GJ, Ransohoff RM, Chensue SW, Raine CS, Lira SA. A novel model of demyelinating encephalomyelitis induced by monocytes and dendritic cells. THE JOURNAL OF IMMUNOLOGY 2007; 177:6871-9. [PMID: 17082601 DOI: 10.4049/jimmunol.177.10.6871] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Local inflammation may be a precipitating event in autoimmune processes. In this study, we demonstrate that regulated influx of monocytes and dendritic cells (DC) into the CNS causes an acute neurological syndrome that results in a demyelinating encephalomyelitis. Expansion of monocytes and DC by conditional expression of Flt3 ligand in animals expressing CCL2 in the CNS promoted parenchymal cell infiltration and ascending paralysis in 100% of the mice within 9 days of Flt3 ligand induction. Depletion of circulating monocytes and DC reduced disease incidence and severity. Unlike the classical models of experimental autoimmune encephalomyelitis, depletion of CD4+ and CD8+ T cells did not affect disease induction. T cells and demyelinating lesions were observed in the CNS at a later stage as a result of organ-specific inflammation. We propose that alterations in the numbers or function of monocytes and DC coupled to dysregulated expression of chemokines in the neural tissues, favors development of CNS autoimmune disease.
Collapse
Affiliation(s)
- Glaucia C Furtado
- Immunobiology Center, Mount Sinai School of Medicine, New York, NY 10029-6574, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2645
|
|
2646
|
|
2647
|
Varol C, Landsman L, Fogg DK, Greenshtein L, Gildor B, Margalit R, Kalchenko V, Geissmann F, Jung S. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. ACTA ACUST UNITED AC 2006; 204:171-80. [PMID: 17190836 PMCID: PMC2118434 DOI: 10.1084/jem.20061011] [Citation(s) in RCA: 494] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mononuclear phagocyte (MP) system is a body-wide macrophage (MPhi) and dendritic cell (DC) network, which contributes to tissue homeostasis, inflammation, and immune defense. The in vivo origins of MPs remain poorly understood. Here, we use an adoptive precursor cell transfer strategy into MP-depleted mice to establish the in vivo differentiation sequence from a recently identified MPhi/DC-restricted bone marrow (BM) precursor (MDP) via BM and blood intermediates to peripheral MPhis and DCs. We show that MDPs are in vivo precursors of BM and blood monocytes. Interestingly, grafted Gr1high "inflammatory" blood monocytes shuttle back to the BM in the absence of inflammation, convert into Gr1low monocytes, and contribute further to MP generation. The grafted monocytes give rise to DCs in the intestinal lamina propria and lung, but not to conventional CD11chigh DCs in the spleen, which develop during homeostasis from MDPs without a monocytic intermediate.
Collapse
Affiliation(s)
- Chen Varol
- Department of Immunology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
2648
|
Abstract
The developmental pathways that lead to the production of antigen-presenting dendritic cells (DCs) are beginning to be understood. These are the last of the pathways of haematopoiesis to be mapped. The existence of many specialized subtypes of DC has complicated this endeavour, as has the need to distinguish the DCs formed in steady state from those produced during an inflammatory response. Here we review studies that lead to the concept that different types of DC develop through different branches of haematopoietic pathways that involve different immediate precursor cells. Furthermore, these studies show that many individual tissues generate their own DCs locally, from a reservoir of immediate DC precursors, rather than depending on a continuous flux of DCs from the bone marrow.
Collapse
Affiliation(s)
- Ken Shortman
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | |
Collapse
|
2649
|
Pradhan S, Genebriera J, Denning WL, Felix K, Elmets CA, Timares L. CD4 T cell-induced, bid-dependent apoptosis of cutaneous dendritic cells regulates T cell expansion and immune responses. THE JOURNAL OF IMMUNOLOGY 2006; 177:5956-67. [PMID: 17056520 PMCID: PMC3466089 DOI: 10.4049/jimmunol.177.9.5956] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The fate of dendritic cells (DCs) after Ag presentation may be DC subset-specific and controlled by many factors. The role of activation-induced apoptosis in regulating DC function is not clear. We investigated the fate of cutaneous DCs (cDCs), specifically Langerhans cells (LCs), and observed that they undergo apoptosis after successful Ag presentation to CD4 T cells. Caspase-specific inhibitors revealed that LC lines use a type II apoptosis pathway in response to CD4 T cells. In support of this, BH3-interacting domain (Bid) protein was present at high levels and specifically cleaved in the presence of Ag-specific T cells. Significant resistance to apoptosis by OT-2 CD4 cells was also observed for Bid knockout (KO) LCs in vitro. To test whether Bid was required to regulate LC function in vivo, we measured contact sensitization and topical immunization responses in Bid KO mice and observed markedly enhanced ear swelling and proliferation responses compared with wild-type mice. Furthermore, when Ag-pulsed Bid KO migratory cDCs were inoculated into wild-type recipients, an increase in both the rate and percentage of expanded OT-2 T cells expressing IFN-gamma was observed. Thus, enhanced Ag presentation function was intrinsic to Bid KO cDCs. Therefore, Bid is an important regulator of LC viability and Ag presentation function.
Collapse
Affiliation(s)
- Sanjay Pradhan
- Department of Dermatology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| | - Joseph Genebriera
- Department of Dermatology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| | - Warren L. Denning
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| | - Kumar Felix
- Department of Dermatology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| | - Craig A. Elmets
- Department of Dermatology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
- The UAB Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| | - Laura Timares
- Department of Dermatology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham AL, 35294, USA
- The UAB Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham AL, 35294, USA
| |
Collapse
|
2650
|
Abstract
PURPOSE OF REVIEW This review focuses on current understanding of molecular mechanisms operating within erythroblastic islands including cell-cell adhesion, regulatory feedback, and central macrophage function. RECENT FINDINGS Erythroblasts express a variety of adhesion molecules and recently two interactions have been identified that appear to be critical for island integrity. Erythroblast macrophage protein, expressed on erythroblasts and macrophages, mediates cell-cell attachments via homophilic binding. Erythroblast intercellular adhesion molecule-4 links erythroblasts to macrophages through interaction with macrophage alphav integrin. In intercellular adhesion molecule-4 knockout mice, erythroblastic islands are markedly reduced, whereas the erythroblast macrophage protein null phenotype is severely anemic and embryonic lethal. Retinoblastoma tumor suppressor (Rb) protein stimulates macrophage differentiation by counteracting inhibition of Id2 on PU.1, a transcription factor that is a crucial regulator of macrophage differentiation. Rb-deficient macrophages do not bind Rb null erythroblasts and the Rb null phenotype is anemic and embryonic lethal. Lastly, extruded nuclei rapidly expose phosphatidylserine on their surface, providing a recognition signal similar to apoptotic cells. SUMMARY Although understanding of molecular mechanisms operating within islands is at an early stage, tantalizing evidence suggests that erythroblastic islands are specialized niches where intercellular interactions in concert with cytokines play critical roles in regulating erythropoiesis.
Collapse
Affiliation(s)
- Joel Anne Chasis
- Life Sciences Division, University of California, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA.
| |
Collapse
|