Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Metab ; 20(1): 61-72, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24930970

ABSTRACT

CD4 T cell activation leads to proliferation and differentiation into effector (Teff) or regulatory (Treg) cells that mediate or control immunity. While each subset prefers distinct glycolytic or oxidative metabolic programs in vitro, requirements and mechanisms that control T cell glucose uptake and metabolism in vivo are uncertain. Despite expression of multiple glucose transporters, Glut1 deficiency selectively impaired metabolism and function of thymocytes and Teff. Resting T cells were normal until activated, when Glut1 deficiency prevented increased glucose uptake and glycolysis, growth, proliferation, and decreased Teff survival and differentiation. Importantly, Glut1 deficiency decreased Teff expansion and the ability to induce inflammatory disease in vivo. Treg cells, in contrast, were enriched in vivo and appeared functionally unaffected and able to suppress Teff, irrespective of Glut1 expression. These data show a selective in vivo requirement for Glut1 in metabolic reprogramming of CD4 T cell activation and Teff expansion and survival.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Glucose Transporter Type 1/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Survival , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
2.
PLoS One ; 8(1): e52087, 2013.
Article in English | MEDLINE | ID: mdl-23300962

ABSTRACT

Two-photon microscopy has enabled the study of individual cell behavior in live animals. Many organs and tissues cannot be studied, especially longitudinally, because they are located too deep, behind bony structures or too close to the lung and heart. Here we report a novel mouse model that allows long-term single cell imaging of many organs. A wide variety of live tissues were successfully engrafted in the pinna of the mouse ear. Many of these engrafted tissues maintained the normal tissue histology. Using the heart and thymus as models, we further demonstrated that the engrafted tissues functioned as would be expected. Combining two-photon microscopy with fluorescent tracers, we successfully visualized the engrafted tissues at the single cell level in live mice over several months. Four dimensional (three-dimensional (3D) plus time) information of individual cells was obtained from this imaging. This model makes long-term high resolution 4D imaging of multiple organs possible.


Subject(s)
Microscopy, Fluorescence, Multiphoton/methods , Myocardium/pathology , Thymus Gland/pathology , Animals , Apoptosis , Ear Auricle/pathology , Electrocardiography , Flow Cytometry , Heart/physiology , Hematopoietic Stem Cell Transplantation , Imaging, Three-Dimensional , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Nude , Models, Animal , Spleen/pathology , T-Lymphocytes/cytology
3.
Int J Radiat Oncol Biol Phys ; 85(4): 1141-8, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23021438

ABSTRACT

PURPOSE: To investigate whether and how insulin-like growth factor 1 (IGF-1) mitigates hematopoietic toxicity after total body irradiation. METHODS AND MATERIALS: BALB/c mice were irradiated with a lethal dose of radiation (7.5 Gy) and treated with IGF-1 at a dose of 100 µg/dose intravenously once a day for 5 consecutive days starting within 1 hour after exposure. Survival and hematopoietic recovery were monitored. The mechanisms by which IGF-1 promotes hematopoietic recovery were also studied by use of an in vitro culture system. RESULTS: IGF-1 protected 8 of 20 mice (40%) from lethal irradiation, whereas only 2 of 20 mice (10%) in the saline control group survived for more than 100 days after irradiation. A single dose of IGF-1 (500 µg) was as effective as daily dosing for 5 days. Positive effects were noted even when the initiation of treatment was delayed as long as 6 hours after irradiation. In comparison with the saline control group, treatment with IGF-1 significantly accelerated the recovery of both platelets and red blood cells in peripheral blood, total cell numbers, hematopoietic stem cells, and progenitor cells in the bone marrow when measured at day 14 after irradiation. IGF-1 protected both hematopoietic stem cells and progenitor cells from radiation-induced apoptosis and cell death. In addition, IGF-1 was able to facilitate the proliferation and differentiation of nonirradiated and irradiated hematopoietic progenitor cells. CONCLUSIONS: IGF-1 mitigates radiation-induced hematopoietic toxicity through protecting hematopoietic stem cells and progenitor cells from apoptosis and enhancing proliferation and differentiation of the surviving hematopoietic progenitor cells.


Subject(s)
Hematinics/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Insulin-Like Growth Factor I/pharmacology , Whole-Body Irradiation/adverse effects , Animals , Apoptosis/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/radiation effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Survival/drug effects , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/radiation effects , Hematopoiesis/physiology , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/physiology , Hematopoietic Stem Cells/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation Dosage , Time Factors
4.
Biol Blood Marrow Transplant ; 18(10): 1488-99, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22809867

ABSTRACT

We studied whether allospecific CD4(+) effector memory T cells (T(EM)) could induce graft-versus-host disease (GVHD) using a novel GVHD model induced solely by CD4(+) T cell receptor transgenic TEa cells. Allospecific T(EM) generated in a lymphopenic host bore a typical memory phenotype. Moreover, these cells were able to elicit a faster and more effective proliferative response on challenge with alloantigen in vitro and to mediate "second-set" skin graft rejection in vivo. However, these allospecific T(EM) were unable to induce GVHD. Allospecific T(EM) recipients became tolerant to alloantigen as a result of clonal deletion. Even though allospecific T(EM) were able to respond to alloantigen initially, the expansion of these cells and inflammatory cytokine production during GVHD were dramatically decreased. The inability of allospecific T(EM) to sustain the alloresponse may be a result of enhanced activation-induced cell death. These observations provide insight into how allospecific CD4(+) T(EM) respond to alloantigen during GVHD and underscore the fundamental differences in alloresponses mediated by allospecific T(EM) in graft rejection and GVHD settings.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/prevention & control , Isoantigens/immunology , Skin Transplantation/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Death/immunology , Clonal Anergy , Clonal Deletion , Cytokines/biosynthesis , Cytokines/immunology , Female , Graft vs Host Disease/immunology , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/cytology
5.
Blood ; 119(26): 6344-53, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22596261

ABSTRACT

A major challenge in allogeneic hematopoietic cell transplantation is how to transfer T-cell immunity without causing graft-versus-host disease (GVHD). Effector memory T cells (CD62L(-)) are a cell subset that can potentially address this challenge because they do not induce GVHD. Here, we investigated how CD62L(-) T cells contributed to phenotypic and functional T-cell reconstitution after transplantation. On transfer into allogeneic recipients, CD62L(-) T cells were activated and expressed multiple cytokines and cytotoxic molecules. CD62L(-) T cells were able to deplete host radioresistant T cells and facilitate hematopoietic engraftment, resulting in enhanced de novo T-cell regeneration. Enhanced functional immune reconstitution was demonstrated in CD62L(-) T-cell recipients using a tumor and an influenza virus challenge model. Even though CD62L(-) T cells are able to respond to alloantigens and deplete host radioresistant immune cells in GVHD recipients, alloreactive CD62L(-) T cells lost the reactivity over time and were eventually tolerant to alloantigens as a result of prolonged antigen exposure, suggesting a mechanism by which CD62L(-) T cells were able to eliminate host resistance without causing GVHD. These data further highlight the unique characteristics of CD62L(-) T cells and their potential applications in clinical hematopoietic cell transplantation.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cell Transplantation , Immunologic Memory/immunology , L-Selectin/metabolism , T-Lymphocytes/physiology , Transplantation Tolerance/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Female , Immunity, Innate/immunology , Immunocompromised Host/immunology , Immunocompromised Host/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
6.
Int J Radiat Biol ; 87(8): 869-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21480768

ABSTRACT

PURPOSE: Radiation and wound combined injury represents a major clinical challenge because of the synergistic interactions that lead to higher morbidity and mortality than either insult would produce singly. The purpose of this study was to develop a mouse ear punch model to study the physiological mechanisms underlying radiation effects on healing wounds. MATERIALS AND METHODS: Surgical wounds were induced by a 2 mm surgical punch in the ear pinnae of MRL/MpJ mice. Photographs of the wounds were taken and the sizes of the ear punch wounds were quantified by image analysis. Local radiation to the ear was delivered by orthovoltage X-ray irradiator using a specially constructed jig that shields the other parts of body. RESULTS: Using this model, we demonstrated that local radiation to the wound area significantly delayed the healing of ear punch wounds in a dose-dependent fashion. The addition of sublethal whole body irradiation (7 Gy) further delayed the healing of ear punch wounds. These results were replicated in C57BL/6 mice; however, wound healing in MRL/MpJ mice was accelerated. CONCLUSIONS: These data indicate that the mouse ear punch model is a valuable model to study radiation and wound combined injury.


Subject(s)
Disease Models, Animal , Ear/injuries , Ear/radiation effects , Wound Healing/radiation effects , Wounds, Penetrating/physiopathology , Animals , Ear/physiopathology , Female , Mice , Mice, Inbred C57BL , Radiation Dosage
7.
PLoS One ; 5(6): e11316, 2010 Jun 28.
Article in English | MEDLINE | ID: mdl-20596257

ABSTRACT

The interaction between stromal cell-derived factor-1 (SDF-1) with CXCR4 chemokine receptors plays an important role in hematopoiesis following hematopoietic stem cell transplantation. We examined the efficacy of post transplant administration of a specific CXCR4 antagonist (AMD3100) in improving animal survival and in enhancing donor hematopoietic cell engraftment using a congeneic mouse transplantation model. AMD3100 was administered subcutaneously at 5 mg/kg body weight 3 times a week beginning at day +2 post-transplant. Post-transplant administration of AMD3100 significantly improves animal survival. AMD3100 reduces pro-inflammatory cytokine/chemokine production. Furthermore, post transplant administration of AMD3100 selectively enhances donor cell engraftment and promotes recovery of all donor cell lineages (myeloid cells, T and B lymphocytes, erythrocytes and platelets). This enhancement results from a combined effect of increased marrow niche availability and greater cell division induced by AMD3100. Our studies shed new lights into the biological roles of SDF-1/CXCR4 interaction in hematopoietic stem cell engraftment following transplantation and in transplant-related mortality. Our results indicate that AMD3100 provides a novel approach for enhancing hematological recovery following transplantation, and will likely benefit patients undergoing transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Heterocyclic Compounds/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Cyclams , Cytokines/biosynthesis , Hematopoietic Stem Cells/metabolism , Mice
8.
PLoS One ; 5(6): e11056, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20585403

ABSTRACT

Medications that can mitigate against radiation injury are limited. In this study, we investigated the ability of recombinant human growth hormone (rhGH) to mitigate against radiation injury in mice and nonhuman primates. BALB/c mice were irradiated with 7.5 Gy and treated post-irradiation with rhGH intravenously at a once daily dose of 20 microg/dose for 35 days. rhGH protected 17 out of 28 mice (60.7%) from lethal irradiation while only 3 out of 28 mice (10.7%) survived in the saline control group. A shorter course of 5 days of rhGH post-irradiation produced similar results. Compared with the saline control group, treatment with rhGH on irradiated BALB/c mice significantly accelerated overall hematopoietic recovery. Specifically, the recovery of total white cells, CD4 and CD8 T cell subsets, B cells, NK cells and especially platelets post radiation exposure were significantly accelerated in the rhGH-treated mice. Moreover, treatment with rhGH increased the frequency of hematopoietic stem/progenitor cells as measured by flow cytometry and colony forming unit assays in bone marrow harvested at day 14 after irradiation, suggesting the effects of rhGH are at the hematopoietic stem/progenitor level. rhGH mediated the hematopoietic effects primarily through their niches. Similar data with rhGH were also observed following 2 Gy sublethal irradiation of nonhuman primates. Our data demonstrate that rhGH promotes hematopoietic engraftment and immune recovery post the exposure of ionizing radiation and mitigates against the mortality from lethal irradiation even when administered after exposure.


Subject(s)
Growth Hormone/administration & dosage , Radiation Injuries, Experimental/prevention & control , Animals , Apoptosis/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Primates , Recombinant Proteins/administration & dosage , T-Lymphocyte Subsets
9.
Blood ; 109(10): 4164-7, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17264295

ABSTRACT

CALGB 9511 used pegaspargase (PEG-ASP) in lieu of the native enzyme. The aim was to compare differences in overall survival (OS) and disease-free survival (DFS) between patients who did and did not achieve asparagine depletion, defined by enzyme levels greater than 0.03 U/mL plasma for 14 consecutive days after at least 1 of 4 planned PEG-ASP administrations. Samples were available from 85 eligible patients. On univariate analyses, the 22 patients who did not achieve asparagine depletion had inferior OS (P = .002; hazard ratio [HR] = 2.37; 95% CI = 1.38-4.09) and DFS (P = .012; HR = 2.21; 95% CI = 1.19-4.13). After adjusting for age, performance status, leukocyte count, and karyotype in a proportional hazards model, both the OS and DFS HRs decreased to 1.8 (P = .056; 95% CI = 1.0-3.2 and P = .084; 95% CI = 0.9-3.6, respectively). We conclude that effective asparagine depletion with PEG-ASP is feasible as part of an intensive multiagent therapeutic regimen in adult acute lymphoblastic leukemia and appears associated with improved outcomes.


Subject(s)
Asparaginase/therapeutic use , Asparagine/blood , Polyethylene Glycols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Feasibility Studies , Humans , Immunophenotyping , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Survival Analysis , Treatment Outcome
10.
Blood ; 109(7): 3115-23, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17148592

ABSTRACT

Several groups, including our own, have independently demonstrated that effector memory T cells from non-alloantigen-primed donors do not cause graft-versus-host disease (GVHD). In the current study, we further investigated whether this approach could be extended to all memory T cells, and we studied the underlying mechanisms. Neither total memory T cells nor purified central memory T cells were able to induce GVHD. Memory T cells were at least 3-log less potent than bulk T cells in mediating GVHD. As expected, memory T cells failed to elicit cytotoxicity and proliferated poorly against alloantigens in standard 5-day mixed-lymphocyte cultures. However, the proliferative responses of memory T cells were more comparable with those of bulk and naive T cells when the culture time was shortened. Moreover, the frequencies of IL-2-secreting cells measured by 42-hour enzyme-linked immunosorbent spot (ELISPOT) assay were similar among naive, memory, and bulk T cells. These data indicated that memory T cells are able to respond to alloantigens initially but fail to develop to full potential. The abortive immune response, which was mediated by non-alloantigen-specific memory T cells in response to alloantigens, may explain why memory T cells from unprimed and non-alloantigen-primed donors could not induce GVHD.


Subject(s)
Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , Animals , Animals, Congenic , CD4-Positive T-Lymphocytes/immunology , Female , Hemocyanins/immunology , In Vitro Techniques , Isoantigens/administration & dosage , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Stem Cell Transplantation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...