251
|
Brunner S, Huber BC, Weinberger T, Vallaster M, Wollenweber T, Gerbitz A, Hacker M, Franz WM. Migration of bone marrow-derived cells and improved perfusion after treatment with erythropoietin in a murine model of myocardial infarction. J Cell Mol Med 2012; 16:152-9. [PMID: 21362129 PMCID: PMC3823101 DOI: 10.1111/j.1582-4934.2011.01286.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Erythropoietin (EPO) was shown to have protective effects after myocardial infarction (MI) by neovascularization and antiapoptotic mechanisms. Beside direct receptor-dependent mechanisms, mobilization and homing of bone marrow-derived cells (BMCs) may play a pivotal role in this regard. In this study, we intended to track different subpopulations of BMCs and to assess serially myocardial perfusion changes in EPO-treated mice after MI. To allow tracking of BMCs, we used a chimeric mouse model. Therefore, mice (C57BL/6J) were sublethally irradiated, and bone marrow (BM) from green fluorescent protein transgenic mice was transplanted. Ten weeks later coronary artery ligation was performed to induce MI. EPO was injected for 3 days with a total dose of 5000 IU/kg. Subpopulations (CD31, c-kit, CXCR-4 and Sca-1) of EGFP+ cells were studied in peripheral blood, bone marrow and hearts by flow cytometry. Myocardial perfusion was serially investigated in vivo by pinhole single-photon emission computed tomography (SPECT) at days 6 and 30 after MI. EPO-treated animals revealed an enhanced mobilization of BMCs into peripheral blood. The numbers of these cells in BM remained unchanged. Homing of all BMCs subpopulations to the ischaemic myocardium was significantly increased in EPO-treated mice. Among the investigated subpopulations, EPO predominantly affected migration of CXCR-4+ (4.3-fold increase). Repetitively SPECT analyses revealed a reduction of perfusion defects after EPO treatment over time. Our study shows that EPO treatment after MI enhances the migration capacity of BMCs into ischaemic tissue, which may attribute to an improved perfusion and reduced size of infarction, respectively.
Collapse
Affiliation(s)
- Stefan Brunner
- Medical Department I, Klinikum der Universität München, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
252
|
Claus C, Riether C, Schürch C, Matter MS, Hilmenyuk T, Ochsenbein AF. CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. Cancer Res 2012; 72:3664-76. [PMID: 22628427 DOI: 10.1158/0008-5472.can-11-2791] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signaling of the TNF receptor superfamily member CD27 activates costimulatory pathways to elicit T- and B-cell responses. CD27 signaling is regulated by the expression of its ligand CD70 on subsets of dendritic cells and lymphocytes. Here, we analyzed the role of the CD27-CD70 interaction in the immunologic control of solid tumors in Cd27-deficient mice. In tumor-bearing wild-type mice, the CD27-CD70 interaction increased the frequency of regulatory T cells (Tregs), reduced tumor-specific T-cell responses, increased angiogenesis, and promoted tumor growth. CD27 signaling reduced apoptosis of Tregs in vivo and induced CD4(+) effector T cells (Teffs) to produce interleukin-2, a key survival factor for Tregs. Consequently, the frequency of Tregs and growth of solid tumors were reduced in Cd27-deficient mice or in wild-type mice treated with monoclonal antibody to block CD27 signaling. Our findings, therefore, provide a novel mechanism by which the adaptive immune system enhances tumor growth and may offer an attractive strategy to treat solid tumors.
Collapse
Affiliation(s)
- Christina Claus
- Tumor Immunology, Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
253
|
Depletion of GR-1-Positive Cells Is Associated with Reduced Neutrophil Inflammation and Astrocyte Reactivity after Experimental Intracerebral Hemorrhage. Transl Stroke Res 2012; 3:147-54. [PMID: 24323867 DOI: 10.1007/s12975-012-0184-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/16/2012] [Accepted: 04/19/2012] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhage (ICH) is the stroke subtype with the highest mortality and morbidity with 25% of patients dying within the first 48 h and a high incidence of poor outcomes. Because of high early mortality rates, an understanding of acute brain injury mechanisms is essential. In this study, we have investigated the putative role of acute inflammation in brain injury after experimental ICH. We depleted GR-1(+) cells in mice by intraperitoneal administration of anti-GR-1 antibody or normal rat serum (control). We then induced ICH by infusion of autologous whole blood into the striatum and compared functional outcome and brain injury markers between the two groups. We found that administration of anti-GR-1 antibody led to a profound decrease in circulating GR-1(+) cells (1.5 ± 0.34% vs. 50.3 ± 8.3% of CD45(+) cells, p ≤ 0.01) and that brain neutrophils decreased by approximately 50% (p ≤ 0.05). We observed a reduction in astrocyte immunoreactivity in the GR-1(+) cell-depleted group (p ≤ 0.05). Conversely, we did not find attenuation of brain edema or differences in behavioral deficits between the two groups. In summary, our results are promising and suggest that larger studies or different neutrophil manipulations may produce greater attenuation of injury after ICH.
Collapse
|
254
|
Yurchenko E, Shio MT, Huang TC, Da Silva Martins M, Szyf M, Levings MK, Olivier M, Piccirillo CA. Inflammation-driven reprogramming of CD4+ Foxp3+ regulatory T cells into pathogenic Th1/Th17 T effectors is abrogated by mTOR inhibition in vivo. PLoS One 2012; 7:e35572. [PMID: 22545118 PMCID: PMC3335853 DOI: 10.1371/journal.pone.0035572] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 03/20/2012] [Indexed: 11/19/2022] Open
Abstract
While natural CD4(+)Foxp3(+) regulatory T (nT(REG)) cells have long been viewed as a stable and distinct lineage that is committed to suppressive functions in vivo, recent evidence supporting this notion remains highly controversial. We sought to determine whether Foxp3 expression and the nT(REG) cell phenotype are stable in vivo and modulated by the inflammatory microenvironment. Here, we show that Foxp3(+) nT(REG) cells from thymic or peripheral lymphoid organs reveal extensive functional plasticity in vivo. We show that nT(REG) cells readily lose Foxp3 expression, destabilizing their phenotype, in turn, enabling them to reprogram into Th1 and Th17 effector cells. nT(REG) cell reprogramming is a characteristic of the entire Foxp3(+) nT(REG) population and the stable Foxp3(NEG) T(REG) cell phenotype is associated with a methylated foxp3 promoter. The extent of nT(REG) cell reprogramming is modulated by the presence of effector T cell-mediated signals, and occurs independently of variation in IL-2 production in vivo. Moreover, the gut microenvironment or parasitic infection favours the reprogramming of Foxp3(+) T(REG) cells into effector T cells and promotes host immunity. IL-17 is predominantly produced by reprogrammed Foxp3(+) nT(REG) cells, and precedes Foxp3 down-regulation, a process accentuated in mesenteric sites. Lastly, mTOR inhibition with the immunosuppressive drug, rapamycin, stabilizes Foxp3 expression in T(REG) cells and strongly inhibits IL-17 but not RORγt expression in reprogrammed Foxp3(-) T(REG) cells. Overall, inflammatory signals modulate mTOR signalling and influence the stability of the Foxp3(+) nT(REG) cell phenotype.
Collapse
Affiliation(s)
- Ekaterina Yurchenko
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Marina T. Shio
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Tony C. Huang
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Maria Da Silva Martins
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Megan K. Levings
- Child and Family Research Institute, Department of Surgery, University of British Columbia, Vancouver, British Colombia, Canada
| | - Martin Olivier
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Ciriaco A. Piccirillo
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
- FOCIS Center of Excellence, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
255
|
Abutbul S, Shapiro J, Szaingurten-Solodkin I, Levy N, Carmy Y, Baron R, Jung S, Monsonego A. TGF-β signaling through SMAD2/3 induces the quiescent microglial phenotype within the CNS environment. Glia 2012; 60:1160-71. [PMID: 22511296 DOI: 10.1002/glia.22343] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/22/2012] [Indexed: 12/16/2022]
Abstract
Microglia are myeloid-derived cells that colonize the central nervous system (CNS) at early stages of development and constitute up to 20% of the glial populations throughout life. While extensive progress has been recently made in identifying the cellular origin of microglia, the mechanism whereby the cells acquire the unique ramified and quiescent phenotype within the CNS milieu remains unknown. Here, we show that upon co-culturing of either CD117(+) /Lin(-) hematopoietic progenitors or CD11c(+) bone marrow derived cells with organotypic hippocampal slices or primary glia, the cells acquire a ramified morphology concomitant with reduced levels of CD86, MHCII, and CD11c and up-regulation of the microglial cell-surface proteins CX(3) CR1 and Iba-1. We further demonstrate that the transforming growth factor beta (TGF-β) signaling pathway via SMAD2/3 phosphorylation is essential for both primary microglia and myeloid-derived cells in order to acquire their quiescent phenotype. Our study suggests that the abundant expression of TGF-β within the CNS during development and various inflammatory processes plays a key role in promoting the quiescent phenotype of microglia and may thus serve as a target for therapeutic strategies aimed at modulating the function of microglia in neurodegenerative diseases such as Alzheimer's and prion.
Collapse
Affiliation(s)
- Shai Abutbul
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | | |
Collapse
|
256
|
Connexin-43 in the osteogenic BM niche regulates its cellular composition and the bidirectional traffic of hematopoietic stem cells and progenitors. Blood 2012; 119:5144-54. [PMID: 22498741 DOI: 10.1182/blood-2011-07-368506] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Connexin-43 (Cx43), a gap junction protein involved in control of cell proliferation, differentiation and migration, has been suggested to have a role in hematopoiesis. Cx43 is highly expressed in osteoblasts and osteogenic progenitors (OB/P). To elucidate the biologic function of Cx43 in the hematopoietic microenvironment (HM) and its influence in hematopoietic stem cell (HSC) activity, we studied the hematopoietic function in an in vivo model of constitutive deficiency of Cx43 in OB/P. The deficiency of Cx43 in OB/P cells does not impair the steady state hematopoiesis, but disrupts the directional trafficking of HSC/progenitors (Ps) between the bone marrow (BM) and peripheral blood (PB). OB/P Cx43 is a crucial positive regulator of transstromal migration and homing of both HSCs and progenitors in an irradiated microenvironment. However, OB/P Cx43 deficiency in nonmyeloablated animals does not result in a homing defect but induces increased endosteal lodging and decreased mobilization of HSC/Ps associated with proliferation and expansion of Cxcl12-secreting mesenchymal/osteolineage cells in the BM HM in vivo. Cx43 controls the cellular content of the BM osteogenic microenvironment and is required for homing of HSC/Ps in myeloablated animals.
Collapse
|
257
|
Gerbitz A, Sukumar M, Helm F, Wilke A, Friese C, Fahrenwaldt C, Lehmann FM, Loddenkemper C, Kammertoens T, Mautner J, Schmitt CA, Blankenstein T, Bornkamm GW. Stromal interferon-γ signaling and cross-presentation are required to eliminate antigen-loss variants of B cell lymphomas in mice. PLoS One 2012; 7:e34552. [PMID: 22479645 PMCID: PMC3316708 DOI: 10.1371/journal.pone.0034552] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/05/2012] [Indexed: 11/19/2022] Open
Abstract
To study mechanisms of T cell-mediated rejection of B cell lymphomas, we developed a murine lymphoma model wherein three potential rejection antigens, human c-MYC, chicken ovalbumin (OVA), and GFP are expressed. After transfer into wild-type mice 60–70% of systemically growing lymphomas expressing all three antigens were rejected; lymphomas expressing only human c-MYC protein were not rejected. OVA expressing lymphomas were infiltrated by T cells, showed MHC class I and II upregulation, and lost antigen expression, indicating immune escape. In contrast to wild-type recipients, 80–100% of STAT1-, IFN-γ-, or IFN-γ receptor-deficient recipients died of lymphoma, indicating that host IFN-γ signaling is critical for rejection. Lymphomas arising in IFN-γ- and IFN-γ-receptor-deficient mice had invariably lost antigen expression, suggesting that poor overall survival of these recipients was due to inefficient elimination of antigen-negative lymphoma variants. Antigen-dependent eradication of lymphoma cells in wild-type animals was dependent on cross-presentation of antigen by cells of the tumor stroma. These findings provide first evidence for an important role of the tumor stroma in T cell-mediated control of hematologic neoplasias and highlight the importance of incorporating stroma-targeting strategies into future immunotherapeutic approaches.
Collapse
Affiliation(s)
- Armin Gerbitz
- Department of Immunology, Charité Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Wieser F, Wu J, Shen Z, Taylor RN, Sidell N. Retinoic acid suppresses growth of lesions, inhibits peritoneal cytokine secretion, and promotes macrophage differentiation in an immunocompetent mouse model of endometriosis. Fertil Steril 2012; 97:1430-7. [PMID: 22464761 DOI: 10.1016/j.fertnstert.2012.03.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 02/22/2012] [Accepted: 03/05/2012] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To determine the effects of all-trans-retinoic acid (RA) on establishment and growth of endometrial lesions, peritoneal interleukin-6 (IL-6) and macrophage chemotactic factor-1 (MCP-1) concentrations, and CD38, CD11b, and F4/80 expression on peritoneal macrophages in an immunocompetent mouse model of endometriosis. DESIGN Experimental transplantation study using mice. SETTING Academic medical center. ANIMAL(S) C57BL/6 recipient mice and syngeneic green fluorescent protein transgenic (GFP+) mice. INTERVENTION(S) Recipient mice were inoculated with GFP+ minced uterine tissue to induce endometriosis and treated with RA (400 nmol/day) or vehicle for 17 days (3 days before to 14 days after tissue injection). MAIN OUTCOME MEASURE(S) Total number of GFP+ implants in recipient mice, number of implants showing visible blood vessels, total volume of established lesions per mouse, concentrations of IL-6 and MCP-1 in peritoneal fluid, and expression of CD11b, F4/80, and CD38 on peritoneal macrophages. RESULT(S) Retinoic acid treatment for 17 days reduced the number of implants versus controls and decreased the frequency of lesions with vessels. Peritoneal washings in RA-treated animals had lower concentrations of IL-6 and MCP-1 than controls 3 days after endometrial inoculation and lower levels of IL-6 on day 14 after inoculation. Concomitant with these effects on day 14, CD38, CD11b, and F4/80 were higher on macrophages from RA-treated mice versus controls. CONCLUSION(S) The development of endometriotic implants is inhibited by RA. This effect may be caused, at least in part, by reduced IL-6 and MCP-1 production and enhanced differentiation of peritoneal macrophages.
Collapse
Affiliation(s)
- Friedrich Wieser
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
259
|
Trans-differentiation of neural stem cells: a therapeutic mechanism against the radiation induced brain damage. PLoS One 2012; 7:e25936. [PMID: 22347993 PMCID: PMC3277599 DOI: 10.1371/journal.pone.0025936] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 09/13/2011] [Indexed: 01/19/2023] Open
Abstract
Radiation therapy is an indispensable therapeutic modality for various brain diseases. Though endogenous neural stem cells (NSCs) would provide regenerative potential, many patients nevertheless suffer from radiation-induced brain damage. Accordingly, we tested beneficial effects of exogenous NSC supplementation using in vivo mouse models that received whole brain irradiation. Systemic supplementation of primarily cultured mouse fetal NSCs inhibited radiation-induced brain atrophy and thereby preserved brain functions such as short-term memory. Transplanted NSCs migrated to the irradiated brain and differentiated into neurons, astrocytes, or oligodendrocytes. In addition, neurotrophic factors such as NGF were significantly increased in the brain by NSCs, indicating that both paracrine and replacement effects could be the therapeutic mechanisms of NSCs. Interestingly, NSCs also differentiated into brain endothelial cells, which was accompanied by the restoration the cerebral blood flow that was reduced from the irradiation. Inhibition of the VEGF signaling reduced the migration and trans-differentiation of NSCs. Therefore, trans-differentiation of NSCs into brain endothelial cells by the VEGF signaling and the consequential restoration of the cerebral blood flow would also be one of the therapeutic mechanisms of NSCs. In summary, our data demonstrate that exogenous NSC supplementation could prevent radiation-induced functional loss of the brain. Therefore, successful combination of brain radiation therapy and NSC supplementation would provide a highly promising therapeutic option for patients with various brain diseases.
Collapse
|
260
|
Characterization of thrombopoietin (TPO)-responsive progenitor cells in adult mouse bone marrow with in vivo megakaryocyte and erythroid potential. Proc Natl Acad Sci U S A 2012; 109:2364-9. [PMID: 22308484 DOI: 10.1073/pnas.1121385109] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hematopoietic progenitor cells are the progeny of hematopoietic stem cells that coordinate the production of precise numbers of mature blood cells of diverse functional lineages. Identification of cell-surface antigen expression associated with hematopoietic lineage restriction has allowed prospective isolation of progenitor cells with defined hematopoietic potential. To clarify further the cellular origins of megakaryocyte commitment, we assessed the in vitro and in vivo megakaryocyte and platelet potential of defined progenitor populations in the adult mouse bone marrow. We show that megakaryocytes arise from CD150(+) bipotential progenitors that display both platelet- and erythrocyte-producing potential in vivo and that can develop from the Flt3(-) fraction of the pregranulocyte-macrophage population. We define a bipotential erythroid-megakaryocyte progenitor population, the CD150(+)CD9(lo)endoglin(lo) fraction of Lin(-)cKit(+)IL7 receptor alpha(-)FcγRII/III(lo)Sca1(-) cells, which contains the bulk of the megakaryocyte colony-forming capacity of the bone marrow, including bipotential megakaryocyte-erythroid colony-forming capacity, and can generate both erythrocytes and platelets efficiently in vivo. This fraction is distinct from the CD150(+)CD9(hi)endoglin(lo) fraction, which contains bipotential precursors with characteristics of increased megakaryocytic maturation, and the CD150(+)CD9(lo)endoglin(hi) fraction, which contains erythroid lineage-committed cells. Finally, we demonstrate that bipotential erythroid-megakaryocyte progenitor and CD150(+)CD9(hi)endoglin(lo) cells are TPO-responsive and that the latter population specifically expands in the recovery from thrombocytopenia induced by anti-platelet serum.
Collapse
|
261
|
Muench MO, Chen JC, Beyer AI, Fomin ME. Cellular therapies supplement: the peritoneum as an ectopic site of hematopoiesis following in utero transplantation. Transfusion 2012; 51 Suppl 4:106S-117S. [PMID: 22074621 DOI: 10.1111/j.1537-2995.2011.03373.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND In utero transplantation (IUT) has the potential to treat birth defects early before full development of the immune system. Relatively small grafts, which are not matched for major histocompatibility antigens, can be delivered even before onset of disease symptoms. IUT of hematopoietic stem cells is usually performed via intraperitoneal injection, yet the fate of donor cells in the peritoneal cavity is not fully understood. We review our recent work and present new data demonstrating that the peritoneum can be a site of ectopic hematopoiesis with implications for IUT and immune tolerance induction. STUDY DESIGN AND METHODS Haplogeneic and allogeneic fetal transplants were performed in mice and engraftment tracked by flow cytometry. Immune tolerance was studied by mixed lymphocyte reactions and skin transplantation. Adult syngeneic murine transplants and xenogeneic human into immunodeficient mouse transplants were performed to follow hematopoietic retention in the peritoneum and engraftment of the marrow. RESULTS Although most transplanted cells rapidly clear the peritoneum, hematopoietic cells and cells with the phenotype of hematopoietic precursors can remain in the peritoneal cavity for months after transplant. The presence of donor cells in the peritoneum can contribute to donor-specific tolerance, but sufficient peripheral blood chimerism is required to ensure acceptance of donor skin grafts. CONCLUSION Ectopic hematopoiesis and the survival of stem cells in the peritoneum offer the possibility of better using the peritoneal cavity to delivery stem cells and foster the development of immune tolerance to alloantigens or other foreign antigens.
Collapse
Affiliation(s)
- Marcus O Muench
- Blood Systems Research Institute, San Francisco, California 94118, USA.
| | | | | | | |
Collapse
|
262
|
Yurchenko E, Levings MK, Piccirillo CA. CD4+Foxp3+ regulatory T cells suppress γδ T-cell effector functions in a model of T-cell-induced mucosal inflammation. Eur J Immunol 2011; 41:3455-66. [DOI: 10.1002/eji.201141814] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 09/09/2011] [Accepted: 09/20/2011] [Indexed: 01/05/2023]
|
263
|
Liang Y, Silva KA, Kennedy V, Sundberg JP. Comparisons of mouse models for hair follicle reconstitution. Exp Dermatol 2011; 20:1011-5. [DOI: 10.1111/j.1600-0625.2011.01366.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
264
|
Josefsson EC, James C, Henley KJ, Debrincat MA, Rogers KL, Dowling MR, White MJ, Kruse EA, Lane RM, Ellis S, Nurden P, Mason KD, O'Reilly LA, Roberts AW, Metcalf D, Huang DCS, Kile BT. Megakaryocytes possess a functional intrinsic apoptosis pathway that must be restrained to survive and produce platelets. ACTA ACUST UNITED AC 2011; 208:2017-31. [PMID: 21911424 PMCID: PMC3182050 DOI: 10.1084/jem.20110750] [Citation(s) in RCA: 148] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Deletion of Bak and Bax, the effectors of mitochondrial apoptosis, does not affect platelet production, however, loss of prosurvival Bcl-xL results in megakaryocyte apoptosis and failure of platelet shedding. It is believed that megakaryocytes undergo a specialized form of apoptosis to shed platelets. Conversely, a range of pathophysiological insults, including chemotherapy, are thought to cause thrombocytopenia by inducing the apoptotic death of megakaryocytes and their progenitors. To resolve this paradox, we generated mice with hematopoietic- or megakaryocyte-specific deletions of the essential mediators of apoptosis, Bak and Bax. We found that platelet production was unperturbed. In stark contrast, deletion of the prosurvival protein Bcl-xL resulted in megakaryocyte apoptosis and a failure of platelet shedding. This could be rescued by deletion of Bak and Bax. We examined the effect on megakaryocytes of three agents that activate the intrinsic apoptosis pathway in other cell types: etoposide, staurosporine, and the BH3 mimetic ABT-737. All three triggered mitochondrial damage, caspase activation, and cell death. Deletion of Bak and Bax rendered megakaryocytes resistant to etoposide and ABT-737. In vivo, mice with a Bak−/− Bax−/− hematopoietic system were protected against thrombocytopenia induced by the chemotherapeutic agent carboplatin. Thus, megakaryocytes do not activate the intrinsic pathway to generate platelets; rather, the opposite is true: they must restrain it to survive and progress safely through proplatelet formation and platelet shedding.
Collapse
Affiliation(s)
- Emma C Josefsson
- Molecular Medicine Division, Cancer and Hematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Croker BA, O'Donnell JA, Nowell CJ, Metcalf D, Dewson G, Campbell KJ, Rogers KL, Hu Y, Smyth GK, Zhang JG, White M, Lackovic K, Cengia LH, O'Reilly LA, Bouillet P, Cory S, Strasser A, Roberts AW. Fas-mediated neutrophil apoptosis is accelerated by Bid, Bak, and Bax and inhibited by Bcl-2 and Mcl-1. Proc Natl Acad Sci U S A 2011; 108:13135-40. [PMID: 21768356 PMCID: PMC3156212 DOI: 10.1073/pnas.1110358108] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During immune responses, neutrophils must integrate survival and death signals from multiple sources to regulate their lifespan. Signals that activate either the Bcl-2- or death receptor-regulated apoptosis pathways can provide powerful stimuli for neutrophils to undergo cell death, but whether they act cooperatively in parallel or directly cross-talk in neutrophils is not known. Previous studies suggested that Bcl-2 family proteins are not required for Fas-induced cell death in neutrophils, but did not examine whether they could modulate its rapid onset. By monitoring the rate of change in neutrophil viability associated with activation of the Fas-triggered death receptor pathway using real-time cell imaging, we show that the Bcl-2-related proteins Bid, Bax, and Bak accelerate neutrophil apoptosis but are not essential for cell death. Increased Bcl-2 or Mcl-1 expression prevents efficient induction of apoptosis by Fas stimulation indicating that the Bcl-2-regulated apoptosis pathway can directly interfere with Fas-triggered apoptosis. Fas has been shown to initiate NFκB activation and gene transcription in cell lines, however gene transcription is not altered in Fas-activated Bid(-/-) neutrophils, indicating that apoptosis occurs independently of gene transcription in neutrophils. The specification of kinetics of neutrophil apoptosis by Bid impacts on the magnitude of neutrophil IL-1β production, implicating a functional role for the Bcl-2-regulated pathway in controlling neutrophil responses to FasL. These data demonstrate that the intrinsic apoptosis pathway directly controls the kinetics of Fas-triggered apoptosis in neutrophils.
Collapse
Affiliation(s)
- Ben A. Croker
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Joanne A. O'Donnell
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Cameron J. Nowell
- Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Royal Melbourne Hospital, Parkville, Victoria 3052, Australia; and
| | - Donald Metcalf
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Grant Dewson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Kirsteen J. Campbell
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Kelly L. Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Yifang Hu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
- Mathematics and Statistics, and
| | - Gordon K. Smyth
- Departments of Medical Biology and
- Mathematics and Statistics, and
- the Faculty of Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Michael White
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Kurt Lackovic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Louise H. Cengia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Lorraine A. O'Reilly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Suzanne Cory
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
| | - Andrew W. Roberts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Departments of Medical Biology and
- the Faculty of Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
266
|
Ruddell A, Harrell MI, Furuya M, Kirschbaum SB, Iritani BM. B lymphocytes promote lymphogenous metastasis of lymphoma and melanoma. Neoplasia 2011; 13:748-57. [PMID: 21847366 PMCID: PMC3156665 DOI: 10.1593/neo.11756] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 06/13/2011] [Accepted: 06/15/2011] [Indexed: 12/17/2022]
Abstract
The prognosis of patients with many types of cancers correlates with the degree of metastasis to regional lymph nodes (LNs) and vital organs. However, the mechanisms and route of cancer cell metastasis are still unclear. Previous studies determined that B-cell accumulation in tumor-draining LNs (TDLNs) induces lymphatic sinus growth (lymphangiogenesis) and increases lymph flow, which could actively promote tumor dissemination through the lymphatic system. Using young Eµ-c-Myc mice that feature LN B-cell expansion as hosts for tumor transplants, we show that subcutaneously implanted lymphomas or melanomas preferentially spread to TDLNs over non-TDLNs, thus demonstrating that these tumors initially metastasize through lymphatic rather than through hematogenous routes. In addition, the rate and amount of tumor dissemination is greater in Eµ-c-Myc mice versus wild-type hosts, which correlates with LN B-cell accumulation and lymphangiogenesis in Eµ-c-Myc hosts. The increased lymphatic dissemination in Eµ-c-Myc hosts is further associated with rapid hematogenous tumor spread of subcutaneously implanted lymphomas, suggesting that TDLN metastasis secondarily drives lymphoma spread to distant organs. In contrast, after intravenous tumor cell injection, spleen metastasis of lymphoma cells or lung metastasis of melanoma cells is similar in Eµ-c-Myc and wild-type hosts. These studies demonstrate that the effect of Eµ-c-Myc hosts to promote metastasis is limited to the lymphatic route of dissemination. TDLN B-cell accumulation, in association with lymphangiogenesis and increased lymph flow, thus significantly contributes to dissemination of lymphomas and solid tumors, providing new targets for therapeutic intervention to block metastasis.
Collapse
Affiliation(s)
- Alanna Ruddell
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | |
Collapse
|
267
|
Erg is required for self-renewal of hematopoietic stem cells during stress hematopoiesis in mice. Blood 2011; 118:2454-61. [PMID: 21673349 DOI: 10.1182/blood-2011-03-344739] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are rare residents of the bone marrow responsible for the lifelong production of blood cells. Regulation of the balance between HSC self-renewal and differentiation is central to hematopoiesis, allowing precisely regulated generation of mature blood cells at steady state and expanded production at times of rapid need, as well as maintaining ongoing stem cell capacity. Erg, a member of the Ets family of transcription factors, is deregulated in cancers; and although Erg is known to be required for regulation of adult HSCs, its precise role has not been defined. We show here that, although heterozygosity for functional Erg is sufficient for adequate steady-state HSC maintenance, Erg(+/Mld2) mutant mice exhibit impaired HSC self-renewal after bone marrow transplantation or during recovery from myelotoxic stress. Moreover, although mice functionally compromised for either Erg or Mpl, the receptor for thrombopoietin, a key regulator of HSC quiescence, maintained sufficient HSC activity to sustain hematopoiesis, Mpl(-/-) Erg(+/Mld2) compound mutant mice displayed exacerbated stem cell deficiencies and bone marrow failure. Thus, Erg is a critical regulator of adult HSCs, essential for maintaining self-renewal at times of high HSC cycling.
Collapse
|
268
|
Campisi L, Soudja SM, Cazareth J, Bassand D, Lazzari A, Brau F, Narni-Mancinelli E, Glaichenhaus N, Geissmann F, Lauvau G. Splenic CD8α⁺ dendritic cells undergo rapid programming by cytosolic bacteria and inflammation to induce protective CD8⁺ T-cell memory. Eur J Immunol 2011; 41:1594-605. [PMID: 21469106 DOI: 10.1002/eji.201041036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 01/08/2011] [Accepted: 03/09/2011] [Indexed: 11/09/2022]
Abstract
Memory CD8(+) T lymphocytes are critical effector cells of the adaptive immune system mediating long-lived pathogen-specific protective immunity. Three signals - antigen, costimulation and inflammation - orchestrate optimal CD8(+) T-cell priming and differentiation into effector and memory cells and shape T-cell functional fate and ability to protect against challenge infections. While among the conventional spleen DCs (cDCs), the CD8α(+) but not the CD8α(-) cDCs most efficiently mediate CD8(+) T-cell priming, it is unclear which subset, irrespective of their capacity to process MHC class I-associated antigens, is most efficient at inducing naïve CD8(+) T-cell differentiation into pathogen-specific protective memory cells in vivo. Moreover, the origin of the required signals is still unclear. Using mice infected with the intracellular bacterium Listeria monocytogenes, we show that splenic CD8α(+) cDCs become endowed with all functional features to optimally prime protective memory CD8(+) T cells in vivo within only a few hours post-immunization. Such programming requires both cytosolic signals resulting from bacterial invasion of the host cells and extracellular inflammatory mediators. Thus, these data designate these cells as the best candidates to facilitate the development of cell-based vaccine therapy.
Collapse
Affiliation(s)
- Laura Campisi
- Institut National de Santé et de Recherche Médicale Unité 924, Groupe Avenir, Valbonne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
269
|
Egen JG, Rothfuchs AG, Feng CG, Horwitz MA, Sher A, Germain RN. Intravital imaging reveals limited antigen presentation and T cell effector function in mycobacterial granulomas. Immunity 2011; 34:807-19. [PMID: 21596592 PMCID: PMC3164316 DOI: 10.1016/j.immuni.2011.03.022] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 01/18/2011] [Accepted: 03/02/2011] [Indexed: 01/18/2023]
Abstract
Cell-mediated adaptive immunity is critical for host defense, but little is known about T cell behavior during delivery of effector function. Here we investigate relationships among antigen presentation, T cell motility, and local production of effector cytokines by CD4+ T cells within hepatic granulomas triggered by Bacille Calmette-Guérin or Mycobacterium tuberculosis. At steady-state, only small fractions of mycobacteria-specific T cells showed antigen-induced migration arrest within granulomas, resulting in low-level, polarized secretion of cytokines. However, exogenous antigen elicited rapid arrest and robust cytokine production by the vast majority of effector T cells. These findings suggest that limited antigen presentation and/or recognition within granulomas evoke a muted T cell response drawing on only a fraction of the host's potential effector capacity. Our results provide new insights into the regulation of host-protective functions, especially how antigen availability influences T cell dynamics and, in turn, effector T cell function during chronic infection.
Collapse
Affiliation(s)
- Jackson G. Egen
- Lymphocyte Biology Section, Laboratory of Immunology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Antonio Gigliotti Rothfuchs
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Carl G. Feng
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California-Los Angeles School of Medicine, Los Angeles, CA USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immunology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
- Program in Systems Immunology and Infectious Disease Modeling, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
270
|
Stemberger S, Jamnig A, Stefanova N, Lepperdinger G, Reindl M, Wenning GK. Mesenchymal stem cells in a transgenic mouse model of multiple system atrophy: immunomodulation and neuroprotection. PLoS One 2011; 6:e19808. [PMID: 21625635 PMCID: PMC3097217 DOI: 10.1371/journal.pone.0019808] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 04/04/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) are currently strong candidates for cell-based therapies. They are well known for their differentiation potential and immunoregulatory properties and have been proven to be potentially effective in the treatment of a large variety of diseases, including neurodegenerative disorders. Currently there is no treatment that provides consistent long-term benefits for patients with multiple system atrophy (MSA), a fatal late onset α-synucleinopathy. Principally neuroprotective or regenerative strategies, including cell-based therapies, represent a powerful approach for treating MSA. In this study we investigated the efficacy of intravenously applied MSCs in terms of behavioural improvement, neuroprotection and modulation of neuroinflammation in the (PLP)-αsynuclein (αSYN) MSA model. METHODOLOGY/PRINCIPAL FINDINGS MSCs were intravenously applied in aged (PLP)-αSYN transgenic mice. Behavioural analyses, defining fine motor coordination and balance capabilities as well as stride length analysis, were performed to measure behavioural outcome. Neuroprotection was assessed by quantifying TH neurons in the substantia nigra pars compacta (SNc). MSC treatment on neuroinflammation was analysed by cytokine measurements (IL-1α, IL-2, IL-4, IL-5, IL-6, IL-10, IL-17, GM-CSF, INFγ, MCP-1, TGF-β1, TNF-α) in brain lysates together with immunohistochemistry for T-cells and microglia. Four weeks post MSC treatment we observed neuroprotection in the SNc, as well as downregulation of cytokines involved in neuroinflammation. However, there was no behavioural improvement after MSC application. CONCLUSIONS/SIGNIFICANCE To our knowledge this is the first experimental approach of MSC treatment in a transgenic MSA mouse model. Our data suggest that intravenously infused MSCs have a potent effect on immunomodulation and neuroprotection. Our data warrant further studies to elucidate the efficacy of systemically administered MSCs in transgenic MSA models.
Collapse
Affiliation(s)
- Sylvia Stemberger
- Divison of Clinical Neurobiology, Department of Neurology, Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | | | | | |
Collapse
|
271
|
Schwickert TA, Victora GD, Fooksman DR, Kamphorst AO, Mugnier MR, Gitlin AD, Dustin ML, Nussenzweig MC. A dynamic T cell-limited checkpoint regulates affinity-dependent B cell entry into the germinal center. ACTA ACUST UNITED AC 2011; 208:1243-52. [PMID: 21576382 PMCID: PMC3173244 DOI: 10.1084/jem.20102477] [Citation(s) in RCA: 325] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Entry into the germinal center requires antigen-bearing B cells to compete for cognate T cell help at the T–B border. The germinal center (GC) reaction is essential for the generation of the somatically hypermutated, high-affinity antibodies that mediate adaptive immunity. Entry into the GC is limited to a small number of B cell clones; however, the process by which this limited number of clones is selected is unclear. In this study, we demonstrate that low-affinity B cells intrinsically capable of seeding a GC reaction fail to expand and become activated in the presence of higher-affinity B cells even before GC coalescence. Live multiphoton imaging shows that selection is based on the amount of peptide–major histocompatibility complex (pMHC) presented to cognate T cells within clusters of responding B and T cells at the T–B border. We propose a model in which T cell help is restricted to the B cells with the highest amounts of pMHC, thus allowing for a dynamic affinity threshold to be imposed on antigen-binding B cells.
Collapse
Affiliation(s)
- Tanja A Schwickert
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
272
|
Berretta F, St-Pierre J, Piccirillo CA, Stevenson MM. IL-2 Contributes to Maintaining a Balance between CD4+Foxp3+ Regulatory T Cells and Effector CD4+ T Cells Required for Immune Control of Blood-Stage Malaria Infection. THE JOURNAL OF IMMUNOLOGY 2011; 186:4862-71. [DOI: 10.4049/jimmunol.1003777] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
273
|
Norton MT, Fortner KA, Oppenheimer KH, Bonney EA. Evidence that CD8 T-cell homeostasis and function remain intact during murine pregnancy. Immunology 2011; 131:426-37. [PMID: 20553337 DOI: 10.1111/j.1365-2567.2010.03316.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Evolving models of immune tolerance have challenged the view that the response of the maternal immune system to environmental or fetal antigens must be suppressed or deviated. CD8 T cells play a central role in the immune response to viruses and intracellular pathogens so the maintenance of both the number and function of these cells is critical to protect both the mother and fetus. We show that the numbers of maternal CD8 T cells in both the spleen and the uterine draining lymph nodes are transiently increased at mid-gestation and this correlates with enhanced CD8 T-cell proliferation and an increased relative expression of both pro-survival and pro-apoptotic molecules. In transgenic mice bearing T-cell antigen receptors specific for the male HY or allo-antigens, the transgenic CD8 T cells retain the ability to proliferate and function during pregnancy. Moreover, anti-HY T-cell receptor transgenic mice have normal numbers of male pups despite the presence of CD8 T cells at the maternal-fetal interface. These data suggest that pregnancy is a dynamic state in which CD8 T-cell turnover is increased while the function and ending size of the CD8 T-cell compartment are maintained.
Collapse
Affiliation(s)
- Michelle T Norton
- University of Vermont College of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
274
|
Sanos SL, Nowak J, Fallet M, Bajenoff M. Stromal Cell Networks Regulate Thymocyte Migration and Dendritic Cell Behavior in the Thymus. THE JOURNAL OF IMMUNOLOGY 2011; 186:2835-41. [DOI: 10.4049/jimmunol.1003563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
275
|
Taoudi S, Bee T, Hilton A, Knezevic K, Scott J, Willson TA, Collin C, Thomas T, Voss AK, Kile BT, Alexander WS, Pimanda JE, Hilton DJ. ERG dependence distinguishes developmental control of hematopoietic stem cell maintenance from hematopoietic specification. Genes Dev 2011; 25:251-62. [PMID: 21245161 DOI: 10.1101/gad.2009211] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development. We show that, from the fetal period, ERG acts as a direct upstream regulator of Gata2 and Runx1 gene activity. Without ERG, physiological HSC maintenance fails, leading to the rapid exhaustion of definitive hematopoiesis.
Collapse
Affiliation(s)
- Samir Taoudi
- Molecular Medicine Division, The Walter and Eliza Institute of Medical Research, Melbourne, Parkville, Victoria 3052, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Huber BC, Brunner S, Segeth A, Nathan P, Fischer R, Zaruba MM, Vallaster M, Theiss HD, David R, Gerbitz A, Franz WM. Parathyroid hormone is a DPP-IV inhibitor and increases SDF-1-driven homing of CXCR4(+) stem cells into the ischaemic heart. Cardiovasc Res 2011; 90:529-37. [PMID: 21245057 DOI: 10.1093/cvr/cvr014] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIMS Parathyroid hormone (PTH) has been shown to promote stem cell mobilization into peripheral blood. Moreover, PTH treatment after myocardial infarction (MI) improved survival and myocardial function associated with enhanced homing of bone marrow-derived stem cells (BMCs). To unravel the molecular mechanisms of PTH-mediated stem cell trafficking, we analysed wild-type (wt) and green fluorescent protein (GFP)-transgenic mice after MI with respect to the pivotal stromal cell-derived factor-1 (SDF-1)/chemokine receptor type 4 (CXCR4) axis. METHODS AND RESULTS WT and GFP-transgenic mice (C57BL/6J) were infarcted by coronary artery ligation and PTH (80 μg/kg/day) was injected for 6 days afterwards. Number of BMCs was analysed by flow cytometry. SDF-1 protein levels and activity of dipeptidyl peptidase-IV (DPP-IV) were investigated by ELISA and activity assay. Functional analyses were performed at day 30 after MI. PTH-treated animals revealed an enhanced homing of CXCR4(+) BMCs associated with an increased protein level of the corresponding homing factor SDF-1 in the ischaemic heart. In vitro and in vivo, PTH inhibited the activity of DPP-IV, which cleaves and inactivates SDF-1. Functionally, PTH significantly improved myocardial function after MI. Both stem cell homing as well as functional recovery were reversed by the CXCR4 antagonist AMD3100. CONCLUSION In summary, PTH is a DPP-IV inhibitor leading to an increased cardiac SDF-1 level, which enhances recruitment of CXCR4(+) BMCs into the ischaemic heart associated with attenuated ischaemic cardiomyopathy. Since PTH is already clinically used our findings may have direct impact on the initiation of studies in patients with ischaemic disorders.
Collapse
Affiliation(s)
- Bruno C Huber
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
277
|
Mosimann C, Kaufman CK, Li P, Pugach EK, Tamplin OJ, Zon LI. Ubiquitous transgene expression and Cre-based recombination driven by the ubiquitin promoter in zebrafish. Development 2011; 138:169-77. [PMID: 21138979 DOI: 10.1242/dev.059345] [Citation(s) in RCA: 330] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Molecular genetics approaches in zebrafish research are hampered by the lack of a ubiquitous transgene driver element that is active at all developmental stages. Here, we report the isolation and characterization of the zebrafish ubiquitin (ubi) promoter, which drives constitutive transgene expression during all developmental stages and analyzed adult organs. Notably, ubi expresses in all blood cell lineages, and we demonstrate the application of ubi-driven fluorophore transgenics in hematopoietic transplantation experiments to assess true multilineage potential of engrafted cells. We further generated transgenic zebrafish that express ubiquitous 4-hydroxytamoxifen-controlled Cre recombinase activity from a ubi:cre(ERt2) transgene, as well as ubi:loxP-EGFP-loxP-mCherry (ubi:Switch) transgenics and show their use as a constitutive fluorescent lineage tracing reagent. The ubi promoter and the transgenic lines presented here thus provide a broad resource and important advancement for transgenic applications in zebrafish.
Collapse
|
278
|
Grazia TJ, Plenter RJ, Lepper HM, Victorino F, Miyamoto SD, Crossno JT, Pietra BA, Gill RG, Zamora MR. Prolongation of cardiac allograft survival by a novel population of autologous CD117+ bone marrow-derived progenitor cells. Am J Transplant 2011; 11:34-44. [PMID: 21114653 PMCID: PMC3059253 DOI: 10.1111/j.1600-6143.2010.03335.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Autologous CD117(+) progenitor cells (PC) have been successfully utilized in myocardial infarction and ischemic injury, potentially through the replacement/repair of damaged vascular endothelium. To date, such cells have not been used to enhance solid organ transplant outcome. In this study, we determined whether autologous bone marrow-derived CD117(+) PC could benefit cardiac allograft survival, possibly by replacing donor vascular cells. Autologous, positively selected CD117(+) PC were administered posttransplantation and allografts were assessed for acute rejection. Although significant generation of recipient vascular cell chimerism was not observed, transferred PC disseminated both to the allograft and to peripheral lymphoid tissues and facilitated a significant, dose-dependent prolongation of allograft survival. While CD117(+) PC dramatically inhibited alloreactive T cell proliferation in vitro, this property did not differ from nonprotective CD117(-) bone marrow populations. In vivo, CD117(+) PC did not significantly inhibit T cell alloreactivity or increase peripheral regulatory T cell numbers. Thus, rather than inhibiting adaptive immunity to the allograft, CD117(+) PC may play a cytoprotective role in prolonging graft survival. Importantly, autologous CD117(+) PC appear to be distinct from bone marrow-derived mesenchymal stem cells (MSC) previously used to prolong allograft survival. As such, autologous CD117(+) PC represent a novel cellular therapy for promoting allograft survival.
Collapse
Affiliation(s)
- T J Grazia
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
279
|
Andersen DC, Kliem A, Schrøder HD, Jensen CH. Newly formed skeletal muscle fibers are prone to false positive immunostaining by rabbit antibodies. Acta Histochem 2011; 113:68-71. [PMID: 19767062 DOI: 10.1016/j.acthis.2009.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2009] [Revised: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 11/16/2022]
Abstract
Reports on muscle biology and regeneration often implicate immuno(cyto/histo)chemical protein characterization using rabbit polyclonal antibodies. In this study we demonstrate that newly formed myofibers are especially prone to false positive staining by rabbit antibodies and this unwanted staining is only recognized (1) by a negative muscle tissue control that does not harbor the protein to be examined (fx. from knockout mouse) or (2) by use of a nonsense rabbit antibody that has been prepared in the same way as the antibody of interest. However, many muscle immuno(cyto/histo)chemical studies only rely on controls that reveal non-specific binding by the secondary antibody and neglect that the primary rabbit antibody itself may cause false positive staining of the muscle. We suggest that reliable immuno-based protein detection in newly formed muscle fibers at least requires a nonsense rabbit antibody and optimally a negative muscle/cell control.
Collapse
Affiliation(s)
- Ditte C Andersen
- Department of Immunology and Microbiology, University of Southern Denmark, Odense C, Denmark
| | | | | | | |
Collapse
|
280
|
The dynamics of macrophage infiltration into the arterial wall during atherosclerotic lesion development in low-density lipoprotein receptor knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:413-22. [PMID: 21224078 DOI: 10.1016/j.ajpath.2010.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 07/23/2010] [Accepted: 09/02/2010] [Indexed: 01/25/2023]
Abstract
Atherosclerosis is a progressive disease in which macrophages play an essential role. Macrophage infiltration into the arterial wall induces the development of an early atherosclerotic lesion. However, the dynamics of macrophage infiltration into the arterial wall during lesion progression remain poorly understood. In this study, low-density lipoprotein receptor knockout mice were fed a Western-type diet for 3, 6, 9, and 12 weeks to induce the formation of atherosclerotic lesions with different degrees of complexity. Subsequently, these mice underwent transplantation with bone marrow-overexpressing enhanced green fluorescent protein to track donor-derived cells, including macrophages. After 8 weeks of Western-type diet feeding after transplantation, macrophage infiltration was evaluated by immunohistochemical staining of donor-derived macrophages (enhanced green fluorescent protein-positive F4/80(+)) in the aortic roots. We found that the growth of pre-existing initial lesions was mainly caused by continued recruitment of donor-derived macrophages into the arterial wall. Interestingly, macrophage infiltration into pre-existing more advanced lesions was largely impaired, likely because of the formation of fibrous caps. In addition, interference with the expression of macrophage ATP-binding cassette transporter 1, an ATP-binding cassette transporter involved in cellular cholesterol efflux and macrophage recruitment into tissues, affects the infiltration of macrophages into pre-existing early lesions but not into advanced lesions. In conclusion, our data suggest that the dynamics of macrophage infiltration into the arterial wall vary greatly during atherogenesis and, thus, may affect the efficiency of pharmaceutical interventions aimed at targeting macrophage infiltration into the arterial wall.
Collapse
|
281
|
Declining lymphoid progenitor fitness promotes aging-associated leukemogenesis. Proc Natl Acad Sci U S A 2010; 107:21713-8. [PMID: 21098275 DOI: 10.1073/pnas.1005486107] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aging is associated with the functional decline of cells, tissues, and organs. At the same time, age is the single most important prognostic factor in the development of most human cancers, including chronic myelogenous and acute lymphoblastic leukemias initiated by Bcr-Abl oncogenic translocations. Prevailing paradigms attribute the association between aging and cancers to the accumulation of oncogenic mutations over time, because the accrual of oncogenic events is thought to be the rate-limiting step in initiation and progression of cancers. Conversely, aging-associated functional decline caused by both cell-autonomous and non-cell-autonomous mechanisms is likely to reduce the fitness of stem and progenitor cell populations. This reduction in fitness should be conducive for increased selection of oncogenic mutations that can at least partially alleviate fitness defects, thereby promoting the initiation of cancers. We tested this hypothesis using mouse hematopoietic models. Our studies indicate that the dramatic decline in the fitness of aged B-lymphopoiesis coincides with altered receptor-associated kinase signaling. We further show that Bcr-Abl provides a much greater competitive advantage to old B-lymphoid progenitors compared with young progenitors, coinciding with restored kinase signaling pathways, and that this enhanced competitive advantage translates into increased promotion of Bcr-Abl-driven leukemias. Moreover, impairing IL-7-mediated signaling is sufficient to promote selection for Bcr-Abl-expressing B progenitors. These studies support an unappreciated causative link between aging and cancer: increased selection of oncogenic mutations as a result of age-dependent alterations of the fitness landscape.
Collapse
|
282
|
Victora GD, Schwickert TA, Fooksman DR, Kamphorst AO, Meyer-Hermann M, Dustin ML, Nussenzweig MC. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 2010; 143:592-605. [PMID: 21074050 PMCID: PMC3035939 DOI: 10.1016/j.cell.2010.10.032] [Citation(s) in RCA: 934] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 09/08/2010] [Accepted: 10/21/2010] [Indexed: 02/07/2023]
Abstract
The germinal center (GC) reaction produces high-affinity antibodies by random mutation and selective clonal expansion of B cells with high-affinity receptors. The mechanism by which B cells are selected remains unclear, as does the role of the two anatomically defined areas of the GC, light zone (LZ) and dark zone (DZ). We combined a transgenic photoactivatable fluorescent protein tracer with multiphoton laser-scanning microscopy and flow cytometry to examine anatomically defined LZ and DZ B cells and GC selection. We find that B cell division is restricted to the DZ, with a net vector of B cell movement from the DZ to the LZ. The decision to return to the DZ and undergo clonal expansion is controlled by T helper cells in the GC LZ, which discern between LZ B cells based on the amount of antigen captured and presented. Thus, T cell help, and not direct competition for antigen, is the limiting factor in GC selection.
Collapse
Affiliation(s)
- Gabriel D. Victora
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
- Martin and Helen Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Tanja A. Schwickert
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - David R. Fooksman
- Martin and Helen Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Alice O. Kamphorst
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Michael Meyer-Hermann
- Department Systems Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, D-38124, Braunschweig, Germany
| | - Michael L. Dustin
- Martin and Helen Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Michel C. Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
283
|
Quantifying subcellular distribution of fluorescent fusion proteins in cells migrating within tissues. Immunol Cell Biol 2010; 89:549-57. [PMID: 20956985 DOI: 10.1038/icb.2010.122] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The movement of proteins within cells can provide dynamic indications of cell signaling and cell polarity, but methods are needed to track and quantify subcellular protein movement within tissue environments. Here we present a semiautomated approach to quantify subcellular protein location for hundreds of migrating cells within intact living tissue using retrovirally expressed fluorescent fusion proteins and time-lapse two-photon microscopy of intact thymic lobes. We have validated the method using GFP-PKCζ, a marker for cell polarity, and LAT-GFP, a marker for T-cell receptor signaling, and have related the asymmetric distribution of these proteins to the direction and speed of cell migration. These approaches could be readily adapted to other fluorescent fusion proteins, tissues and biological questions.
Collapse
|
284
|
Multipotential hematopoietic blast colony-forming cells exhibit delays in self-generation and lineage commitment. Proc Natl Acad Sci U S A 2010; 107:16257-61. [PMID: 20805490 DOI: 10.1073/pnas.1011881107] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Murine hematopoietic blast colony-forming cells (BL-CFCs) are able to generate up to 30,000 progeny blast cells within 10 d in agar cultures. Contained in these populations are large numbers of lineage-committed progenitor cells in the granulocytic and macrophage lineages. Sequential analyses of blast colonies revealed that self-generation of BL-CFCs occurs but is surprisingly late in clonal expansion, as is the emergence of progenitor cells committed to megakaryocytic and eosinophil lineages. Self-generating BL-CFCs were highly enriched in lineage(-) Kit(+) Sca1(+) CD34(-) Flt3R(-) populations, and colonies generated by such cells contained colony-forming units-spleen and formed erythroid and lymphoid progeny in vivo. The data suggest the existence of a hierarchical structure in BL-CFC populations with at least a subset being cells assayable as colony-forming units-spleen. Because BL-CFCs can self-generate and are able to generate lymphoid and myeloid populations, BL-CFCs appear to be ideal cells in which to analyze the processes of self-generation and lineage commitment in clonal in vitro cultures.
Collapse
|
285
|
Wang M, Takeda K, Shiraishi Y, Okamoto M, Dakhama A, Joetham A, Gelfand EW. Peanut-induced intestinal allergy is mediated through a mast cell-IgE-FcepsilonRI-IL-13 pathway. J Allergy Clin Immunol 2010; 126:306-16, 316.e1-12. [PMID: 20624645 PMCID: PMC2917491 DOI: 10.1016/j.jaci.2010.05.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 04/02/2010] [Accepted: 05/17/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND Although implicated in the disease, the specific contributions of FcepsilonRI and IL-13 to the pathogenesis of peanut-induced intestinal allergy are not well defined. OBJECTIVES We sought to determine the contributions of FcepsilonRI, IL-13, and mast cells to the development of intestinal mucosal responses in a murine model of peanut-induced intestinal allergy. METHODS Sensitized wild-type (WT), FcepsilonRI-deficient (FcepsilonRI(-/-)), and mast cell-deficient (Kit(W-sh/W-sh)) mice received peanut orally every day for 1 week. Bone marrow-derived mast cells (BMMCs) from WT, FcepsilonRI(-/-), IL-4(-/-), IL-13(-/-), and IL-4/IL-13(-/-) mice were differentiated and transferred into WT, FcepsilonRI(-/-), and Kit(W-sh/W-sh) recipients. BMMCs from WT and UBI-GFP/BL6 mice were differentiated and transferred into WT and Kit(W-sh/W-sh) mice. Blockade of IL-13 was achieved by using IL-13 receptor alpha2 (IL-13Ralpha2)-IgG fusion protein. RESULTS FcepsilonRI(-/-) mice showed decreased intestinal inflammation (mast cell and eosinophil numbers) and goblet cell metaplasia and reduced levels of IL4, IL6, IL13, and IL17A mRNA expression in the jejunum. Transfer of WT BMMCs to FcepsilonRI(-/-) recipients restored their ability to develop intestinal allergic responses unlike transfer of FcepsilonRI(-/-), IL-13(-/-), or IL-4/IL-13(-/-) BMMCs. FcepsilonRI(-/-) mice exhibited lower IL-13 levels and treatment of WT mice with IL-13 receptor alpha2 prevented peanut-induced intestinal allergy and inflammation. CONCLUSIONS These data indicate that the development of peanut-induced intestinal allergy is mediated through a mast cell-dependent IgE-FcepsilonRI-IL-13 pathway. Targeting IL-13 might be a potential treatment for IgE-mediated peanut-induced allergic responses in the intestine.
Collapse
Affiliation(s)
- Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo, USA
| | | | | | | | | | | | | |
Collapse
|
286
|
Lau AA, Hannouche H, Rozaklis T, Hassiotis S, Hopwood JJ, Hemsley KM. Allogeneic stem cell transplantation does not improve neurological deficits in mucopolysaccharidosis type IIIA mice. Exp Neurol 2010; 225:445-54. [PMID: 20673764 DOI: 10.1016/j.expneurol.2010.07.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 11/17/2022]
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a neurodegenerative metabolic disorder caused by mutations in the N-sulfoglucosamine sulfohydrolase gene with resultant accumulation of partially degraded heparan sulfate (HS). Whilst allogeneic bone marrow transplantation (BMT) is indicated for several lysosomal storage disorders featuring neurodegeneration, its use in MPS III is highly controversial. Published evidence suggests that BMT does not improve cognitive function in MPS III patients. Despite this, patients continue to be transplanted in some centers. We therefore sought to determine the clinical effectiveness of BMT in a murine model of MPS IIIA. Pre-symptomatic young adult mice pre-conditioned with total body irradiation generated complete and stable donor-type chimerism. Whilst HS-derived disaccharides were reduced by up to 27% in the brain parenchyma, this was insufficient to decrease secondary cholesterol and GM3 ganglioside storage or permit clinical improvement. These results suggest that BMT is ineffective in its unmodified form and should not be considered as a treatment for MPS IIIA children.
Collapse
Affiliation(s)
- Adeline A Lau
- Lysosomal Diseases Research Unit, SA Pathology at the Women's and Children's Hospital, Australia.
| | | | | | | | | | | |
Collapse
|
287
|
Alvarez-Saavedra M, Carrasco L, Sura-Trueba S, Demarchi Aiello V, Walz K, Neto JX, Young JI. Elevated expression of MeCP2 in cardiac and skeletal tissues is detrimental for normal development. Hum Mol Genet 2010; 19:2177-2190. [PMID: 20203171 DOI: 10.1093/hmg/ddq096] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
MeCP2 plays a critical role in interpreting epigenetic signatures that command chromatin conformation and regulation of gene transcription. In spite of MeCP2's ubiquitous expression, its functions have always been considered in the context of brain physiology. In this study, we demonstrate that alterations of the normal pattern of expression of MeCP2 in cardiac and skeletal tissues are detrimental for normal development. Overexpression of MeCP2 in the mouse heart leads to embryonic lethality with cardiac septum hypertrophy and dysregulated expression of MeCP2 in skeletal tissue produces severe malformations. We further show that MeCP2's expression in the heart is developmentally regulated; further suggesting that it plays a key role in regulating transcriptional programs in non-neural tissues.
Collapse
|
288
|
Park DJ, Agarwal A, George JF. Heme oxygenase-1 expression in murine dendritic cell subpopulations: effect on CD8+ dendritic cell differentiation in vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2831-9. [PMID: 20395442 PMCID: PMC2877844 DOI: 10.2353/ajpath.2010.090845] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/18/2010] [Indexed: 11/20/2022]
Abstract
Heme oxygenase-1 (HO-1) is a microsomal enzyme with antioxidant, antiapoptotic, and immunoregulatory functions. We studied the expression of HO-1 by bone marrow-derived dendritic cells (BMDCs) and splenic DC subpopulations under quiescent conditions or following lipopolysaccharide (LPS) stimulation. The kinetics of HO-1 expression by BMDCs depended on the conditions under which they were propagated. Expression of HO-1 in mouse BMDCs in 100 U/ml GM-CSF peaked at 16 hours after LPS treatment and maintained expression for at least 48 hours. But cultures in 800 U/ml granulocyte-macrophage colony-stimulating factor (GM-CSF) showed peak expression by 16 hours that disappeared by 48 hours after LPS stimulation, similar to BMDCs cultured in both 100 U/ml GM-CSF and IL-4 (10 ng/ml). By flow cytometry, a large proportion of CD8(+) splenic DCs strongly expressed HO-1, and this population significantly increased following LPS administration in vivo. In HO-1(-/-) mice, the proportion of splenic CD8(+) DCs was significantly decreased in comparison with HO-1(+/+) mice. In addition, a unique subpopulation of MHC II(-)CD11b(+)CD11c(+) cells was prominent in HO-1(-/-) spleens. Injection of GFP-labeled HO-1(+/+) splenic DC precursors into HO-1(+/+) mice resulted in the generation of GFP(+)CD8(+) DCs in the spleen after 5 days, but GFP(+) CD8(+) DCs failed to appear in HO-1(-/-) spleens. Conversely, GFP(+)HO-1(-/-) splenic cells also generated GFP(+)CD8(+) DCs in HO-1(+/+) mice. These results show that HO-1 is involved in splenic DC differentiation, and/or the homing of CD8(+) splenic DC precursors appears to be dependent on HO-1 expression by the host.
Collapse
Affiliation(s)
- Dong Jun Park
- Department of Medicine, Division of Cardiothoracic Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
289
|
Swearingen KE, Loomis WP, Kehimkar B, Cookson BT, Dovichi NJ. Quantification of green fluorescent protein in cellular supernatant by capillary electrophoresis with laser-induced fluorescence detection for measurement of cell death. Talanta 2010; 81:948-53. [PMID: 20298877 PMCID: PMC2842605 DOI: 10.1016/j.talanta.2010.01.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 01/18/2010] [Accepted: 01/19/2010] [Indexed: 11/24/2022]
Abstract
A common method for quantifying cell death is measuring the concentration of lactate dehydrogenase (LDH) released by cells as their membranes become unstable. In cells expressing green fluorescent protein (GFP), degradation of the cell membrane also results in the release of GFP into the surrounding supernatant. In this study, we used capillary electrophoresis with laser-induced fluorescence detection to measure the levels of GFP in supernatants of UBIGFP/BL6 primary macrophages that had been infected with Salmonella typhimurium, treated with staurosporine, or exposed to H(2)O(2), all known inducers of cell death. We also used a standard LDH assay to measure the release of LDH into supernatants. We observed the rate of cell death quantified by release of GFP and LDH into supernatant to be essentially identical, demonstrating that GFP release is at least as good as an indicator of macrophage cell death as the established LDH release method.
Collapse
Affiliation(s)
| | - Wendy P. Loomis
- Departments of Laboratory Medicine and Microbiology, University of Washington, Box 357110, Seattle, WA 98195-7110
| | - Benjamin Kehimkar
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195-1700
| | - Brad T. Cookson
- Departments of Laboratory Medicine and Microbiology, University of Washington, Box 357110, Seattle, WA 98195-7110
| | - Norman J. Dovichi
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195-1700
| |
Collapse
|
290
|
Britschgi MR, Favre S, Luther SA. CCL21 is sufficient to mediate DC migration, maturation and function in the absence of CCL19. Eur J Immunol 2010; 40:1266-71. [DOI: 10.1002/eji.200939921] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
| | - Stéphanie Favre
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
291
|
Marusyk A, Porter CC, Zaberezhnyy V, DeGregori J. Irradiation selects for p53-deficient hematopoietic progenitors. PLoS Biol 2010; 8:e1000324. [PMID: 20208998 PMCID: PMC2830447 DOI: 10.1371/journal.pbio.1000324] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 01/28/2010] [Indexed: 12/22/2022] Open
Abstract
While disruption of p53 is selectively neutral within non-stressed hematopoiesis, it confers a strong selective advantage upon irradiation, leading to expansion of p53 mutant clones and lymphoma development. Identification and characterization of mutations that drive cancer evolution constitute a major focus of cancer research. Consequently, dominant paradigms attribute the tumorigenic effects of carcinogens in general and ionizing radiation in particular to their direct mutagenic action on genetic loci encoding oncogenes and tumor suppressor genes. However, the effects of irradiation are not limited to genetic loci that encode oncogenes and tumor suppressors, as irradiation induces a multitude of other changes both in the cells and their microenvironment which could potentially affect the selective effects of some oncogenic mutations. P53 is a key tumor suppressor, the loss of which can provide resistance to multiple genotoxic stimuli, including irradiation. Given that p53 null animals develop T-cell lymphomas with high penetrance and that irradiation dramatically accelerates lymphoma development in p53 heterozygous mice, we hypothesized that increased selection for p53-deficient cells contributes to the causal link between irradiation and induction of lymphoid malignancies. We sought to determine whether ionizing irradiation selects for p53-deficient hematopoietic progenitors in vivo using mouse models. We found that p53 disruption does not provide a clear selective advantage within an unstressed hematopoietic system or in previously irradiated BM allowed to recover from irradiation. In contrast, upon irradiation p53 disruption confers a dramatic selective advantage, leading to long-term expansion of p53-deficient clones and to increased lymphoma development. Selection for cells with disrupted p53 appears to be attributable to several factors: protection from acute irradiation-induced ablation of progenitor cells, prevention of irradiation-induced loss of clonogenic capacity for stem and progenitor cells, improved long-term maintenance of progenitor cell fitness, and the disabling/elimination of competing p53 wild-type progenitors. These studies indicate that the carcinogenic effect of ionizing irradiation can in part be explained by increased selection for cells with p53 disruption, which protects progenitor cells both from immediate elimination and from long-term reductions in fitness following irradiation. Cancer progression can be understood through the framework of Darwinian evolution, which involves two major factors: genetic mutation and selection. Random mutations are thought to result in the initiation and phenotypic diversification of tumors, and environmental influences mediate selection for those mutations that increase tumor cell fitness. Since oncogenic mutations are necessary for the development of spontaneous malignancies and since experimental introduction of these mutations often leads to transformation and cancers, the causation of cancers by carcinogens is traditionally attributed to their induction of new mutations that are oncogenic. We instead asked whether selection for oncogenic mutations is affected by ionizing irradiation, an archetypal mutagenic carcinogen, by examining the selective effects of inactivation of the critical tumor suppressor gene p53. While disruption of p53 is selectively neutral in populations of unstressed hematopoietic progenitors, it provides a strong selective advantage upon irradiation. This selection of p53-deficient clones is attributable to protection from irradiation-induced cell death and loss of cellular fitness. Importantly, the selective expansion of irradiated cells bearing p53 disruption is blocked in the presence of non-irradiated wild-type competitors, indicating that the disabling of competing wild-type cells by irradiation is critical for selection of p53-deficient cells. Our results argue that induction of cancers by irradiation involves selection for mutations that confer radioresistance, and suggest that greater focus on how carcinogenic contexts impact on selection is warranted in understanding, preventing and treating cancers.
Collapse
Affiliation(s)
- Andriy Marusyk
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Program in Molecular Biology, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - Christopher C. Porter
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Program in Molecular Biology, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Department of Pediatrics, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
292
|
de Waard V, Bot I, de Jager SCA, Talib S, Egashira K, de Vries MR, Quax PHA, Biessen EAL, van Berkel TJC. Systemic MCP1/CCR2 blockade and leukocyte specific MCP1/CCR2 inhibition affect aortic aneurysm formation differently. Atherosclerosis 2010; 211:84-9. [PMID: 20197192 DOI: 10.1016/j.atherosclerosis.2010.01.042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 11/20/2009] [Accepted: 01/28/2010] [Indexed: 11/16/2022]
Abstract
OBJECTIVE CCR2, the receptor for monocyte chemoattractant protein 1 (MCP1), is involved in atherosclerosis and abdominal aortic aneurysms (AAAs). Here, we explored the potential beneficial blockade of the MCP1/CCR2 pathway. METHODS We applied an AAA model in aging apolipoprotein E deficient mice with pre-existing atherosclerotic lesions. These mice were subjected to two therapeutic strategies. First, a dominant negative form of MCP1 was overexpressed in femoral muscles, resulting in circulating levels of MCP1-7ND (7ND), competing with native MCP1. In the second approach, bone marrow transplantation was performed using bone marrow cells that were infected with a lentiviral construct containing siRNA for CCR2, to specifically inhibit only leukocyte CCR2 expression. RESULTS Both strategies did not influence lesion size of the advanced atherosclerotic plaques. However, 7ND induced a more fibrous plaque phenotype. Yet, surprisingly a trend in increased number and severity of AAA was observed in the 7ND group. Smooth muscle cells in the aneurysm showed decreased phosphorylated signal transducer and activator of transcription five (STAT5, P<0.01) in the 7ND group, which is indicative for a decreased proliferative and migratory (wound healing) response. This presumably resulted in the increased AAA development. In contrast, siRNA-induced inhibition of CCR2 in leukocytes led to a significant inhibition in aneurysm formation. In conclusion, systemic inhibition of the MCP1/CCR2 pathway leads to a fibrous plaque phenotype in the advanced atherosclerotic lesions, but to potential adverse effects on AAA formation, implying that for a beneficial overall therapeutic approach, specific inhibitory targeting of leukocyte CCR2 will be essential.
Collapse
Affiliation(s)
- Vivian de Waard
- Division of Biopharmaceutics, Leiden/Amsterdam Center for Drug Research, Leiden University, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Schwickert TA, Alabyev B, Manser T, Nussenzweig MC. Germinal center reutilization by newly activated B cells. ACTA ACUST UNITED AC 2009; 206:2907-14. [PMID: 19934021 PMCID: PMC2806468 DOI: 10.1084/jem.20091225] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Germinal centers (GCs) are specialized structures in which B lymphocytes undergo clonal expansion, class switch recombination, somatic hypermutation, and affinity maturation. Although these structures were previously thought to contain a limited number of isolated B cell clones, recent in vivo imaging studies revealed that they are in fact dynamic and appear to be open to their environment. We demonstrate that B cells can colonize heterologous GCs. Invasion of primary GCs after subsequent immunization is most efficient when T cell help is shared by the two immune responses; however, it also occurs when the immune responses are entirely unrelated. We conclude that GCs are dynamic anatomical structures that can be reutilized by newly activated B cells during immune responses.
Collapse
Affiliation(s)
- Tanja A Schwickert
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | | |
Collapse
|
294
|
Martin-Pagola A, Pileggi A, Zahr E, Vendrame F, Damaris Molano R, Snowhite I, Ricordi C, Eisenbarth GS, Nakayama M, Pugliese A. Insulin2 gene (Ins2) transcription by NOD bone marrow-derived cells does not influence autoimmune diabetes development in NOD-Ins2 knockout mice. Scand J Immunol 2009; 70:439-46. [PMID: 19874548 DOI: 10.1111/j.1365-3083.2009.02316.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Insulin is a critical autoantigen for the development of autoimmune diabetes in non-obese diabetic (NOD) mice. About 80% of NOD females and 30-40% of NOD males develop diabetes. However, Insulin2 (Ins2) knockout NOD mice develop autoimmune diabetes with complete penetrance in both sexes, at an earlier age, and have stronger autoimmune responses to insulin. The severe diabetes phenotype observed in NOD-Ins2-/- mice suggests that lack of Ins2 expression in the thymus may compromise immunological tolerance to insulin. Insulin is a prototypical tissue specific antigen (TSA) for which tolerance is dependent on expression in thymus and peripheral lymphoid tissues. TSA are naturally expressed by medullary thymic epithelial cells (mTEC), stromal cells in peripheral lymphoid tissues and bone marrow (BM)-derived cells, mainly CD11c(+) dendritic cells. The natural expression of TSA by mTEC and stromal cells has been shown to contribute to self-tolerance. However, it is unclear whether this also applies to BM-derived cells naturally expressing TSA. To address this question, we created BM chimeras and investigated whether reintroducing Ins2 expression solely by NOD BM-derived cells delays diabetes development in NOD-Ins2-/- mice. On follow-up, NOD-Ins2-/- mice receiving Ins2-expressing NOD BM cells developed diabetes at similar rates of those receiving NOD-Ins2-/- BM cells. Diabetes developed in 64% of NOD recipients transplanted with NOD BM and in 47% of NOD mice transplanted with NOD-Ins2-/- BM (P = ns). Thus, NOD-Ins2-/- BM did not worsen diabetes in NOD recipients and Ins2 expression by NOD BM-derived cells did not delay diabetes development in NOD-Ins2-/- mice.
Collapse
Affiliation(s)
- A Martin-Pagola
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
295
|
Filipe-Santos O, Pescher P, Breart B, Lippuner C, Aebischer T, Glaichenhaus N, Späth GF, Bousso P. A dynamic map of antigen recognition by CD4 T cells at the site of Leishmania major infection. Cell Host Microbe 2009; 6:23-33. [PMID: 19616763 DOI: 10.1016/j.chom.2009.04.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 03/13/2009] [Accepted: 05/01/2009] [Indexed: 11/16/2022]
Abstract
CD4 T helper cells play a central role in the control of infection by intracellular parasites. How efficiently pathogen-specific CD4 T cells detect infected cells in vivo is unclear. Here, we employed intravital two-photon imaging to examine the behavior of pathogen-specific CD4 T cells at the site of Leishmania major infection. While activated CD4 T cells enter the inflamed tissue irrespective of their antigen specificity, pathogen-specific T cells preferentially decelerated and accumulated in infected regions of the dermis. Antigen recognition by CD4 T cells was heterogeneous, involving both stable and dynamic contacts with infected phagocytes. However, not all infected cells induced arrest or deceleration of pathogen-specific T cells, and dense clusters of infected cells were poorly accessible to migrating T cells. Thus, disparities in the dynamics of T cell contacts with infected cells and local variation in T cell access to infected cells are important elements of the host-pathogen interplay.
Collapse
Affiliation(s)
- Orchidée Filipe-Santos
- Institut Pasteur, G5 Dynamiques des Réponses Immunes, Inserm U668, Equipe Avenir, Paris F-75724, France
| | | | | | | | | | | | | | | |
Collapse
|
296
|
Huang X, Pierce LJ, Chen GL, Chang KT, Spangrude GJ, Prchal JT. Erythropoietin receptor signaling regulates both erythropoiesis and megakaryopoiesis in vivo. Blood Cells Mol Dis 2009; 44:1-6. [PMID: 19836979 DOI: 10.1016/j.bcmd.2009.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2009] [Accepted: 09/21/2009] [Indexed: 12/11/2022]
Abstract
Transgenic expression of a gain-of-function truncated mouse erythropoietin receptor gene (EpoR) leads to expansion of the HSC pool in response to human erythropoietin (Epo). We have re-examined this observation using a knock-in mouse model, wherein the mouse EpoR gene was replaced in its proper genetic locus by a single copy of either a wild-type human or a polycythemia-inducing truncated human EPOR gene. Bone marrow cells obtained from knock-in mice were transplanted together with competitor bone marrow cells in a model that allows tracking of erythroid, platelet, and leukocyte contributions by each genotype. Secondary transplants were also performed. Stem/progenitor cells were identified phenotypically and isolated for colony-forming assays to evaluate cytokine responsiveness by cells with the wild-type human or truncated human EPOR gene. Augmented Epo signaling increased erythroid repopulation post-transplant as expected, but had no effect on short-term or long-term leukocyte repopulation. However, the wild-type human EPOR knock-in mouse showed decreases in both erythroid and platelet repopulation compared to marrow cells from the mutant human EPOR knock-in mouse or normal B6 animals. These results provide evidence supporting a role for Epo signaling in megakaryopoiesis in vivo and suggest a role for Epo signaling early in hematopoietic development.
Collapse
Affiliation(s)
- Xiaosong Huang
- Department of Pathology, University of Utah, Salt Lake City, UT 84132-2408, USA
| | | | | | | | | | | |
Collapse
|
297
|
Transplanted myogenic progenitor cells express neuronal markers in the CNS and ameliorate disease in Experimental Autoimmune Encephalomyelitis. J Neuroimmunol 2009; 215:73-83. [DOI: 10.1016/j.jneuroim.2009.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Revised: 08/23/2009] [Accepted: 08/24/2009] [Indexed: 12/27/2022]
|
298
|
Chtanova T, Han SJ, Schaeffer M, van Dooren GG, Herzmark P, Striepen B, Robey EA. Dynamics of T cell, antigen-presenting cell, and pathogen interactions during recall responses in the lymph node. Immunity 2009; 31:342-55. [PMID: 19699173 DOI: 10.1016/j.immuni.2009.06.023] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 04/27/2009] [Accepted: 06/05/2009] [Indexed: 10/20/2022]
Abstract
Memory T cells circulate through lymph nodes where they are poised to respond rapidly upon re-exposure to a pathogen; however, the dynamics of memory T cell, antigen-presenting cell, and pathogen interactions during recall responses are largely unknown. We used a mouse model of infection with the intracellular protozoan parasite, Toxoplasma gondii, in conjunction with two-photon microscopy, to address this question. After challenge, memory T cells migrated more rapidly than naive T cells, relocalized toward the subcapsular sinus (SCS) near invaded macrophages, and engaged in prolonged interactions with infected cells. Parasite invasion of T cells occurred by direct transfer of the parasite from the target cell into the T cell and corresponded to an antigen-specific increase in the rate of T cell invasion. Our results provide insight into cellular interactions during recall responses and suggest a mechanism of pathogen subversion of the immune response.
Collapse
Affiliation(s)
- Tatyana Chtanova
- Department of Molecular and Cell Biology, Life Sciences Addition, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | |
Collapse
|
299
|
Marusyk A, Casás-Selves M, Henry CJ, Zaberezhnyy V, Klawitter J, Christians U, DeGregori J. Irradiation alters selection for oncogenic mutations in hematopoietic progenitors. Cancer Res 2009; 69:7262-9. [PMID: 19738065 DOI: 10.1158/0008-5472.can-09-0604] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exposure to ionizing radiation and other DNA-damaging carcinogens is strongly associated with induction of malignancies. Prevailing paradigms attribute this association to the induction of oncogenic mutations, as the incidence of oncogenic events is thought to limit initiation and progression of cancers. On the other hand, random mutagenic and genotoxic effects of irradiation are likely to alter progenitor cell populations and the microenvironment, thus altering the selective effects of oncogenic mutations. Using competitive bone marrow transplantation experiments in mice, we show that ionizing irradiation leads to a persistent decline in the numbers and fitness of hematopoietic stem cells, in part resulting from persistent induction of reactive oxygen species. Previous irradiation dramatically alters the selective effects of some oncogenic mutations, substantially inhibiting clonal expansion and leukemogenesis driven by Bcr-Abl or activated N-Ras oncogenes but enhancing the selection for and leukemogenesis driven by the activated Notch1 mutant ICN. Irradiation-dependent selection for ICN expression occurs in a hematopoietic stem cell-enriched pool, which should facilitate the accumulation of additional oncogenic events at a committed T-progenitor stage critical for formation of T-lymphocytic leukemia stem cells. Enhancement of ICN-driven selection and leukemogenesis by previous irradiation is in part non-cell autonomous, as partial restoration of normal hematopoiesis can reverse these effects of irradiation. These studies show that irradiation substantially alters the adaptive landscape in hematopoietic progenitors and suggest that the causal link between irradiation and carcinogenesis might involve increased selection for particular oncogenic mutations.
Collapse
Affiliation(s)
- Andriy Marusyk
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | | | | | | | | | | | | |
Collapse
|
300
|
B-cell follicle development remodels the conduit system and allows soluble antigen delivery to follicular dendritic cells. Blood 2009; 114:4989-97. [PMID: 19713459 DOI: 10.1182/blood-2009-06-229567] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Afferent lymph is transported throughout lymph nodes (LNs) by the conduit system. Whereas this conduit network is dense in the T-cell zone, it is sparse in B-cell follicles. In this study, we show that this differential organization emerges during lymph node development. Neonatal LNs lack B follicles, but have a developed T-cell zone and a dense conduit network. As new T and B cells enter the developing LN, the conduit network density is maintained in the T, but not the B zone, leading to a profound remodeling of the follicular network that nevertheless maintains its connectivity. In adults, the residual follicular conduits transport soluble antigen to deep regions, where follicular dendritic cells are abundant and appear to replace the fibroblastic reticular cells that enwrap conduits in the T zone. This strategic location correlates with the capacity of the follicular dendritic cells to capture antigen even in the absence of antigen-specific antibodies. Together, these results describe how the stromal organization of the T and B regions of LNs diverges during development, giving rise to distinct antigen transport and delivery modes in the 2 compartments.
Collapse
|