Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Blood ; 121(11): 2144-53, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23305740

ABSTRACT

Donor hematopoietic stem cells (HSCs) can correct T-cell deficiencies in patients with severe combined immunodeficiency by replacing resident thymus cells. However, as those progenitors that naturally migrate to the thymus are not capable of supporting long-term thymopoiesis, a successful transplant is thought to require the ongoing migration of donor progenitors. We previously showed that the forced intrathymic administration of histocompatible HSCs can sustain long-term thymopoiesis in ZAP-70-immunodeficient mice. However, it is not known whether T-cell reconstitution across histocompatibility barriers is modulated by intrathymic vs intravenous administration of HSCs. In the absence of conditioning, long-term thymopoiesis by semiallogeneic progenitors was detected in mice transplanted via the intrathymic, but not the intravenous, route. In intrathymic-transplanted mice, ongoing thymopoiesis was associated with a 10-fold higher level of early thymic progenitors (ETPs). The enhanced reconstitution capacity of these intrathymic-derived ETPs was corroborated by their significantly augmented myeloid lineage potential compared with endogenous ETPs. Notably, though, myeloablative conditioning resulted in a reduced expansion of intrathymic-administered donor ETPs. Thus, in the absence of conditioning, the forced thymic entry of HSCs results in a sustained T-cell development across histocompatibility barriers, highlighting the capacity of the thymus to support cells with long-term renewal potential.


Subject(s)
Cell Differentiation/immunology , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility/physiology , Lymphoid Progenitor Cells/physiology , T-Lymphocytes/physiology , Thymus Gland , Animals , Cells, Cultured , Graft Survival/immunology , Graft Survival/physiology , Hematopoiesis/immunology , Hematopoiesis/physiology , Histocompatibility/immunology , Histocompatibility Testing , Infusions, Intravenous , Lymphoid Progenitor Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Thymus Gland/cytology , Transplantation Conditioning/methods , ZAP-70 Protein-Tyrosine Kinase/deficiency , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology
2.
Oncoimmunology ; 1(5): 600-608, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22934252

ABSTRACT

Vav1 is expressed exclusively in hematopoietic cells and is required for T cell development and activation. Vav1-deficient mice show thymic hypocellularity due to a partial block during thymocyte development at the DN3 stage and between the double positive (DP) and single positive (SP) transition. Vav1 has been shown to play a significant role in several non-hematopoietic tumors but its role in leukemogenesis is unknown. To address this question, we investigated the role of Vav1 in retrovirus-induced T cell leukemogenesis. Infection of Vav1-deficient mice with the Moloney strain of murine leukemia virus (M-MuLV) significantly affected tumor phenotype without modulating tumor incidence or latency. M-MuLV-infected Vav1-deficient mice showed reduced splenomegaly, higher hematocrit levels and hypertrophic thymi. Notably, Vav1-deficient mice with M-MuLV leukemias presented with markedly lower TCRß/CD3 levels, indicating that transformation occurred at an earlier stage of T cell development than in WT mice. Thus, impaired T cell development modulates the outcome of retrovirus-induced T cell leukemias, demonstrating a link between T cell development and T cell leukemogenesis.

3.
J Exp Med ; 208(7): 1403-17, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21708927

ABSTRACT

The multifunctional E4F1 protein was originally discovered as a target of the E1A viral oncoprotein. Growing evidence indicates that E4F1 is involved in key signaling pathways commonly deregulated during cell transformation. In this study, we investigate the influence of E4F1 on tumorigenesis. Wild-type mice injected with fetal liver cells from mice lacking CDKN2A, the gene encoding Ink4a/Arf, developed histiocytic sarcomas (HSs), a tumor originating from the monocytic/macrophagic lineage. Cre-mediated deletion of E4F1 resulted in the death of HS cells and tumor regression in vivo and extended the lifespan of recipient animals. In murine and human HS cell lines, E4F1 inactivation resulted in mitochondrial defects and increased production of reactive oxygen species (ROS) that triggered massive cell death. Notably, these defects of E4F1 depletion were observed in HS cells but not healthy primary macrophages. Short hairpin RNA-mediated depletion of E4F1 induced mitochondrial defects and ROS-mediated death in several human myeloid leukemia cell lines. E4F1 protein is overexpressed in a large subset of human acute myeloid leukemia samples. Together, these data reveal a role for E4F1 in the survival of myeloid leukemic cells and support the notion that targeting E4F1 activities might have therapeutic interest.


Subject(s)
DNA-Binding Proteins/deficiency , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Repressor Proteins/deficiency , Transcription Factors/deficiency , Animals , Autophagy/physiology , Base Sequence , Cell Death/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/metabolism , Histiocytic Sarcoma/pathology , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Oxidative Stress , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Ubiquitin-Protein Ligases
4.
PLoS One ; 5(9): e12659, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20856822

ABSTRACT

BACKGROUND: Lymphopenia results in the proliferation and differentiation of naïve T cells into memory-like cells in the apparent absence of antigenic stimulation. This response, at least in part due to a greater availability of cytokines, is thought to promote anti-self responses. Although potentially autoreactive memory-like CD8(+) T cells generated in a lymphopenic environment are subject to the mechanisms of peripheral tolerance, they can induce autoimmunity in the presence of antigen-specific memory-like CD4(+) T helper cells. METHODOLOGY/PRINCIPAL FINDINGS: Here, we studied the mechanisms underlying CD4 help under lymphopenic conditions in transgenic mice expressing a model antigen in the beta cells of the pancreas. Surprisingly, we found that the self-reactivity mediated by the cooperation of memory-like CD8(+) and CD4(+) T cells was not abrogated by CD40L blockade. In contrast, treatment with anti-IL-2 antibodies inhibited the onset of autoimmunity. IL-2 neutralization prevented the CD4-mediated differentiation of memory-like CD8(+) T cells into pathogenic effectors in response to self-antigen cross-presentation. Furthermore, in the absence of helper cells, induction of IL-2 signaling by an IL-2 immune complex was sufficient to promote memory-like CD8(+) T cell self-reactivity. CONCLUSIONS/SIGNIFICANCE: IL-2 mediates the cooperation of memory-like CD4(+) and CD8(+) T cells in the breakdown of cross-tolerance, resulting in effector cytotoxic T lymphocyte differentiation and the induction of autoimmune disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Immunologic Memory , Interleukin-2/immunology , Lymphopenia/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cross-Priming , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
5.
PLoS Pathog ; 6(6): e1000948, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20548955

ABSTRACT

Antiviral monoclonal antibodies (mAbs) represent promising therapeutics. However, most mAbs-based immunotherapies conducted so far have only considered the blunting of viral propagation and not other possible therapeutic effects independent of virus neutralization, namely the modulation of the endogenous immune response. As induction of long-term antiviral immunity still remains a paramount challenge for treating chronic infections, we have asked here whether neutralizing mAbs can, in addition to blunting viral propagation, exert immunomodulatory effects with protective outcomes. Supporting this idea, we report here that mice infected with the FrCas(E) murine retrovirus on day 8 after birth die of leukemia within 4-5 months and mount a non-protective immune response, whereas those rapidly subjected to short immunotherapy with a neutralizing mAb survive healthy and mount a long-lasting protective antiviral immunity with strong humoral and cellular immune responses. Interestingly, the administered mAb mediates lysis of infected cells through an antibody-dependent cell cytotoxicity (ADCC) mechanism. In addition, it forms immune complexes (ICs) with infected cells that enhance antiviral CTL responses through Fc gammaR-mediated binding to dendritic cells (DCs). Importantly, the endogenous antiviral antibodies generated in mAb-treated mice also display the same properties, allowing containment of viral propagation and enhancement of memory cellular responses after disappearance of the administered mAb. Thus, our data demonstrate that neutralizing antiviral mAbs can act as immunomodulatory agents capable of stimulating a protective immunity lasting long after the end of the treatment. They also show an important role of infected-cells/antibody complexes in the induction and the maintenance of protective immunity through enhancement of both primary and memory antiviral T-cell responses. They also indicate that targeting infected cells, and not just viruses, by antibodies can be crucial for elicitation of efficient, long-lasting antiviral T-cell responses. This must be considered when designing antiviral mAb-based immunotherapies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Immunization, Passive , Retroviridae Infections/immunology , Retroviridae Infections/therapy , Retroviridae/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigen-Antibody Complex , Cell Proliferation , Flow Cytometry , Mice
6.
Neurosci Lett ; 475(3): 179-83, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20371377

ABSTRACT

P300/CBP associated factor (PCAF) acts as an acetyltransferase that acetylates specific lysine residues in histones, thereby remodelling chromatin structure. The possible involvement of PCAF in learning and memory processes or mood disorders was recently assessed by characterizing the behavioural phenotype of PCAF KO mice bred on a CD1 background and revealed short-term memory deficits that evolved with age towards long-term memory alteration and an exaggerated response to stress [10]. PCAF KO mice have been backcrossed on a C57BL/6j strain for 15 generations and we report here the first data regarding their behavioural phenotype. PCAF KO mice bred on a C57 background showed short-term memory deficits in terms of decreased spontaneous alternation and absence of acquisition of a daily changing platform position in the water-maze. Acquisition of a fixed platform location or passive avoidance response was preserved. PCAF KO mice showed no difference with WT C57BL/6j controls in their performances in the forced swimming and light/dark exploration box, suggesting no particular phenotype on anxiety and stress responses. We therefore evidenced marked phenotypic differences in PCAF KO mice depending on the genetic background strain confirming that PCAF histone acetyltransferase is involved lifelong in the chromatin remodelling necessary for memory formation but differentially involved in anxiety and response to stress.


Subject(s)
Anxiety/psychology , Histone Acetyltransferases/genetics , Memory, Short-Term , Stress, Psychological/psychology , p300-CBP Transcription Factors/genetics , Animals , Anxiety/genetics , Avoidance Learning , Female , Histone Acetyltransferases/physiology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Reaction Time , Spatial Behavior , Stress, Psychological/genetics , p300-CBP Transcription Factors/physiology
7.
Blood ; 115(10): 1913-20, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20040762

ABSTRACT

The sustained differentiation of T cells in the thymus cannot be maintained by resident intrathymic (IT) precursors and requires that progenitors be replenished from the bone marrow (BM). In patients with severe combined immunodeficiency (SCID) treated by hematopoietic stem cell transplantation, late T-cell differentiation defects are thought to be due to an insufficient entry of donor BM progenitors into the thymus. Indeed, we find that the intravenous injection of BM progenitors into nonconditioned zeta-chain-associated protein kinase 70 (ZAP-70)-deficient mice with SCID supports short- but not long-term thymopoiesis. Remarkably, we now show that the IT administration of these progenitors produces a significant level of donor-derived thymopoiesis for more than 6 months after transplantation. In contrast to physiologic thymopoiesis, long-term donor thymopoiesis was not due to the continued recruitment of progenitors from the BM. Rather, IT transplantation resulted in the unique generation of a large population of early c-Kit(high) donor precursors within the thymus. These ZAP-70-deficient mice that received an IT transplant had a significantly increased prothymocyte niche compared with their untreated counterparts; this phenotype was associated with the generation of a medulla. Thus, IT administration of BM progenitors results in the filling of an expanded precursor niche and may represent a strategy for enhancing T-cell differentiation in patients with SCID.


Subject(s)
Bone Marrow Transplantation/methods , Bone Marrow Transplantation/physiology , Lymphoid Progenitor Cells/transplantation , Lymphopoiesis/physiology , Thymus Gland/cytology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cells, Cultured , Infusions, Intravenous , Lymphocyte Count , Lymphoid Progenitor Cells/physiology , Lymphopoiesis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cell Niche/cytology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Thymus Gland/physiology , Time Factors , ZAP-70 Protein-Tyrosine Kinase/genetics
8.
J Immunol ; 182(6): 3398-405, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265117

ABSTRACT

Tumor cell-based vaccines are currently used in clinical trails, but they are in general poorly immunogenic because they are composed of cell extracts or apoptotic cells. Live tumor cells should be much better Ags provided that they are properly processed by the host immune system. We show herein that stable expression of a small hairpin RNA for ERK5 (shERK5) decreases ERK5 levels in human and mouse leukemic cells and leads to their elimination by NK cells in vivo. The shERK5 cells show down-regulation of MHC class I expression at the plasma membrane. Accordingly, ectopic activation of the ERK5 pathway induces MHC class I gene expression. Coinjection of shERK5-expressing cells into the peritoneum diminishes survival of engrafted wild-type tumor cells. Moreover, s.c. injection of shERK5-expressing cells strongly diminishes tumor development by wild-type cells. Our results show that shERK5 expression in leukemia cells effectively attenuates their tumor activity and allows their use as a tumor cell-based vaccine.


Subject(s)
Cancer Vaccines/immunology , Gene Knockdown Techniques , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Leukemia L1210/prevention & control , Lymphocyte Activation/immunology , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Jurkat Cells , Killer Cells, Natural/metabolism , Leukemia L1210/enzymology , Leukemia L1210/genetics , Leukemia L1210/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 7/biosynthesis , RNA, Small Interfering/physiology , Signal Transduction/genetics , Signal Transduction/immunology
9.
PLoS One ; 4(2): e4533, 2009.
Article in English | MEDLINE | ID: mdl-19225570

ABSTRACT

Indole derivatives compounds (IDC) are a new class of splicing inhibitors that have a selective action on exonic splicing enhancers (ESE)-dependent activity of individual serine-arginine-rich (SR) proteins. Some of these molecules have been shown to compromise assembly of HIV infectious particles in cell cultures by interfering with the activity of the SR protein SF2/ASF and by subsequently suppressing production of splicing-dependent retroviral accessory proteins. For all replication-competent retroviruses, a limiting requirement for infection and pathogenesis is the expression of the envelope glycoprotein which strictly depends on the host splicing machinery. Here, we have evaluated the efficiency of IDC on an animal model of retroviral pathogenesis using a fully replication-competent retrovirus. In this model, all newborn mice infected with a fully replicative murine leukemia virus (MLV) develop erythroleukemia within 6 to 8 weeks of age. We tested several IDC for their ability to interfere ex vivo with MLV splicing and virus spreading as well as for their protective effect in vivo. We show here that two of these IDC, IDC13 and IDC78, selectively altered splicing-dependent production of the retroviral envelope gene, thus inhibiting early viral replication in vivo, sufficiently to protect mice from MLV-induced pathogenesis. The apparent specificity and clinical safety observed here for both IDC13 and IDC78 strongly support further assessment of inhibitors of SR protein splicing factors as a new class of antiretroviral therapeutic agents.


Subject(s)
Indoles/pharmacology , Leukemia Virus, Murine/pathogenicity , Leukemia, Erythroblastic, Acute/prevention & control , Nuclear Proteins/antagonists & inhibitors , Animals , Animals, Newborn , Leukemia Virus, Murine/genetics , Leukemia, Erythroblastic, Acute/drug therapy , Mice , RNA Splicing , RNA-Binding Proteins , Retroviridae , Serine-Arginine Splicing Factors
10.
Neurobiol Aging ; 30(6): 987-1000, 2009 Jun.
Article in English | MEDLINE | ID: mdl-17977621

ABSTRACT

Cystinosis is a lysosomal storage disorder characterised by progressive cystine accumulation. The causative gene, CTNS, encodes cystinosin, the lysosomal cystine transporter. Neurological deterioration is one of the last symptoms to appear and the least well characterised. Visuospatial memory deficits have been documented in patients. To determine whether the cystinosis mouse model presents similar anomalies, we studied the learning and memory abilities of young and middle-aged Ctns(-/-) mice. We did not detect deficits in young Ctns(-/-) mice. In contrast, spatial reference and working memory deficits were detected in middle-aged Ctns(-/-) mice. Elevated cystine levels were detected in the hippocampus, cerebellum, forebrain and brainstem of all Ctns(-/-) mice, which increased with age and were consistent with the appearance of impairments. Our results strongly suggest that the cystinosis-associated CNS anomalies are due to progressive cystine accumulation. Furthermore, the Ctns(-/-) mice serve as a model to investigate the evolution of these anomalies and test the efficiency of existing and novel treatments to cross the blood-brain barrier and reduce lysosomal cystine levels.


Subject(s)
Aging/metabolism , Brain/metabolism , Cystine/metabolism , Memory Disorders/metabolism , Memory , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution
11.
Methods Mol Biol ; 506: 171-90, 2009.
Article in English | MEDLINE | ID: mdl-19110627

ABSTRACT

The thymus provides a specialized environment allowing the differentiation of T lymphocytes from bone marrow-derived progenitor cells. We and others have demonstrated that gene transfer into distinct thymocyte populations can be obtained, both in vivo and ex vivo, using lentiviral vectors. Here, we describe techniques for intrathymic lentiviral transduction in mice, using a surgical approach wherein the thoracic cavity is exposed as well as a significantly less invasive strategy wherein virions are directly injected through the skin. Moreover, thymocyte differentiation from murine and human progenitors is now feasible in vitro, under conditions wherein the Notch and IL-7 signaling pathways are activated. We describe methods allowing transduction of murine and human progenitors and their subsequent differentiation into more mature thymocytes. Conditions for lentiviral gene transfer into more differentiated human thymocyte subsets are also presented. Optimization of technologies for HIV-based gene transfer into murine and human thymocyte progenitors will advance strategies aimed at modulating T-cell differentiation and function in-vivo; approaches potentially targeting patients with genetic and acquired immunodeficiencies as well as immune-sensitive tumors. Furthermore, this technology will foster the progression of basic research aimed at elucidating molecular aspects of T-cell differentiation in mice and humans.


Subject(s)
Gene Transfer Techniques , Thymus Gland/metabolism , Animals , Antigens, CD34/immunology , Base Sequence , Cell Line , DNA Primers , Humans , Mice , Thymus Gland/cytology , Thymus Gland/immunology , Transduction, Genetic
12.
Mol Ther ; 17(3): 472-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19088703

ABSTRACT

The thymus is the primary site of T-cell development and plays a key role in the induction of self-tolerance. We previously showed that the intrathymic (i.t.) injection of a transgene-expressing lentiviral vector (LV) in mice can result in the correction of a T cell-specific genetic defect. Nevertheless, the efficiency of thymocyte transduction did not exceed 0.1-0.3% and we were unable to detect any thymus transduction in macaques. As such, we initiated studies to assess the capacity of recombinant adeno-associated virus (rAAV) vectors to transduce murine and primate thymic cells. In vivo administration of AAV serotype 2-derived single-stranded AAV (ssAAV) and self-complementary AAV (scAAV) vectors pseudotyped with capsid proteins of serotypes 1, 2, 4, 5, and 8 demonstrated that murine thymus transduction was significantly enhanced by scAAV2/8. Transgene expression was detected in 5% of thymocytes and, notably, transduced cells represented 1% of peripheral T lymphocytes. Moreover, i.t. administration of scAAV2/8 particles in macaques, by endoscopic-mediated guidance, resulted in significant gene transfer. Thus, in healthy animals, where thymic gene transfer does not provide a selective advantage, scAAV2/8 is a unique tool promoting the in situ transduction of thymocytes with the subsequent export of gene-modified lymphocytes to the periphery.


Subject(s)
Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Macaca fascicularis/metabolism , Thymus Gland/metabolism , Transgenes/genetics , Animals , Cell Differentiation , Cell Line , Cell Movement , Dependovirus/classification , Genetic Vectors/pharmacology , Genome, Viral/genetics , Humans , Kinetics , Mice , Phenotype , Thymus Gland/cytology
13.
Blood ; 112(12): 4729-38, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18796630

ABSTRACT

Glucose is a major source of energy for living organisms, and its transport in vertebrates is a universally conserved property. Of all cell lineages, human erythrocytes express the highest level of the Glut1 glucose transporter with more than 200,000 molecules per cell. However, we recently reported that erythrocyte Glut1 expression is a specific trait of vitamin C-deficient mammalian species, comprising only higher primates, guinea pigs, and fruit bats. Here, we show that in all other tested mammalian species, Glut1 was transiently expressed in erythrocytes during the neonatal period. Glut1 was up-regulated during the erythroblast stage of erythroid differentiation and was present on the vast majority of murine red blood cells (RBCs) at birth. Notably though, Glut1 was not induced in adult mice undergoing anemia-induced erythropoiesis, and under these conditions, the up-regulation of a distinct transporter, Glut4, was responsible for an increased glucose transport. Sp3 and Sp1 transcriptions factors have been proposed to regulate Glut1 transcription, and we find that the concomitant repression of Glut1 and induction of Glut4 was associated with a significantly augmented Sp3/Sp1 ratio. Glucose transporter expression patterns in mice and human erythrocytes are therefore distinct. In mice, there is a postnatal switch from Glut1 to Glut4, with Glut4 further up-regulated under anemic conditions.


Subject(s)
Erythropoiesis/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 4/genetics , Anemia/genetics , Anemia/pathology , Animals , Animals, Newborn , Cattle , Dogs , Erythrocytes/metabolism , Erythrocytes/pathology , Exosomes/metabolism , Gene Expression Regulation, Developmental , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Transport/physiology , Rats
14.
Mol Ther ; 16(8): 1372-81, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18578013

ABSTRACT

Cystinosis belongs to a growing class of lysosomal storage disorders (LSDs) caused by defective transmembrane proteins. The causative CTNS gene encodes the lysosomal cystine transporter, cystinosin. Currently the aminothiol cysteamine is the only drug available for reducing cystine storage but this treatment has non-negligible side effects and administration constraints. In this study, for the first time, we report viral vector-mediated CTNS gene transfer and evaluate the feasibility of this strategy as a complementary treatment. Initially, we transduced human CTNS(-/-) fibroblast cell lines and primary murine Ctns(-/-) hepatocyte cultures in vitro and demonstrated that gene transfer can reduce cystine storage. Because of age-related increase in cystine levels, we transduced hepatocytes from young (/=5 months of age) mice. Our in vitro data suggested that the efficiency of correction was age-dependent. We tested these observations in vivo: short-term (1 week) and long-term (4 weeks) CTNS-transduction significantly reduced hepatic cystine levels in young, but not older, Ctns(-/-) mice. Our data provide the proof-of-concept that gene transfer is feasible for correcting defective lysosomal transport, but suggest that, in the case of cystinosis, it could be preventive but not curative in some tissues.


Subject(s)
Amino Acid Transport Systems, Neutral/physiology , Cystine/metabolism , Cystinosis/therapy , Genetic Therapy/methods , Adenoviruses, Canine/genetics , Age Factors , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Cystinosis/genetics , Cystinosis/metabolism , Dogs , Feasibility Studies , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Vectors/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Kupffer Cells/cytology , Kupffer Cells/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission
15.
J Leukoc Biol ; 84(2): 380-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18483203

ABSTRACT

The TNF family member, a proliferation-inducing ligand (APRIL), has been suggested to act as a costimulatory molecule in T cell responses. However, studies addressing this role in vivo are largely lacking. Here, we evaluated the effects of APRIL on physiological T cell responses in vivo. Although receptors for APRIL are expressed on a subset of T cells, neither TCR transgenic (Tg) T cell responses nor endogenous TCR responses were affected by Tg APRIL expression in vivo. Moreover, APRIL did not significantly enhance the induction of T cell lymphomas upon Moloney murine leukemia virus (MLV) infection. This clearly contrasts current belief and indicates that APRIL does not serve a major role in T cell immunity or lymphomagenesis. However, we did observe a strong increase in erythroleukemia formation after MLV inoculation of APRIL Tg mice. Strikingly, this erythroleukemia-facilitating property of APRIL was confirmed using the erythroleukemogenic Friend-MLV. Erythroleukemia in APRIL Tg mice was characterized by low hematocrits and grossly enlarged spleens with an increased percentage of erythroid precursors. Altogether, these results unveil new proerythroleukemogenic properties of APRIL.


Subject(s)
Leukemia, Erythroblastic, Acute/immunology , Leukemia, Erythroblastic, Acute/virology , Lymphoma, T-Cell/physiopathology , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology , Animals , Autoimmunity , Flow Cytometry , Hematocrit , Heterozygote , Homozygote , Lymphoma, T-Cell/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Moloney murine leukemia virus/immunology , Moloney murine leukemia virus/pathogenicity , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Stem Cells/physiology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
16.
Mol Immunol ; 45(12): 3463-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18462800

ABSTRACT

The cancer immunosurveillance hypothesis has found strong experimental support in recent years. It is believed that cytotoxic lymphocytes are important effectors in this process. PKCtheta plays an essential role in proliferation, activation and survival of these cells, but also proliferation and survival of leukemic T cells. In light of this, we tested the role of PKCtheta in T cell leukemia progression by inducing this disease in wild-type (wt) and PKCtheta-deficient mice with moloney-murine leukemia virus (M-MuLV). Leukemic PKCtheta(-/-) and wild-type (wt) mice showed the same profile of leukemic cell types, similar spleen and thymus sizes and comparable hematocrits. In contrast, disease incidence was higher and disease onset more rapid in PKCtheta(-/-) mice. Transfer of leukemic T cells from wt donors into PKCtheta-deficient and wt recipients induced leukemia in 100% and 40% of the mice, respectively. Interestingly, leukemic cells from PKCtheta(-/-) donors induced the disease in only 50% of the PKCtheta-deficient and 10% of the wt recipients. Intravenous injection of low numbers of EL4 cells induced tumors earlier in PKCtheta(-/-) mice. Taken together, our results show that PKCtheta is essential for the immune response to leukemia in mice and raise questions about the chronic treatment of humans with PKCtheta inhibitors.


Subject(s)
Isoenzymes/deficiency , Leukemia/enzymology , Leukemia/immunology , Protein Kinase C/deficiency , Animals , Animals, Newborn , Isoenzymes/metabolism , Leukemia/pathology , Leukemia/virology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Phenotype , Protein Kinase C/metabolism , Protein Kinase C-theta , Survival Analysis
17.
J Virol ; 82(3): 1339-49, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032505

ABSTRACT

Neutralizing monoclonal antibodies (MAbs) are increasingly being considered for blunting human viral infections. However, whether they can also exert indirect effects on endogenous antiviral immune responses has been essentially overlooked. We have recently shown that a short (several-day) period of immunotherapy with the neutralizing 667 MAb of mouse neonates shortly after infection with the lethal FrCas(E) retrovirus not only has an immediate effect on the viral load but also permits an endogenous antiviral immunity to emerge. Even though passive immunotherapy was administered during the particular period of immunocompetence acquisition, the endogenous response eventually arising was protective and persisted long (>1 year) after the MAb has disappeared. As very high levels of anti-FrCas(E) antibodies, predominantly of the immunoglobulin G2a (IgG2a) isotype and showing strong neutralization activity, were found in the sera of MAb-treated mice, it was necessary to address whether this humoral immunity was sufficient on its own to confer full protection against FrCas(E) or whether a cytotoxic T-lymphocyte (CTL) response was also necessary. Using a variety of in vivo assays in young and adult animals previously infected by FrCas(E) and treated by 667, we show here that transient 667 immunotherapy is associated with the emergence of a CTL response against virus-infected cells. This cytotoxic activity is indispensable for long-term antiviral protective immunity, as high neutralizing antibody titers, even enhanced in in vivo CD8(+) cell depletion experiments, cannot prevent the FrCas(E)-induced death of infected/treated mice. Our work may have important therapeutic consequences, as it indicates that a short period of MAb-based immunotherapy conducted at a stage where the immune system is still developing can be associated with the mounting of a functional Th1-type immune response characterized by both CTL and IgG2a-type humoral contributions, the cooperation of which is known to be essential for the containment of chronic infections by a variety of viruses.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Immunization, Passive , Retroviridae Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Animals, Newborn , Antibodies, Viral/immunology , Lymphocyte Depletion , Mice , Neutralization Tests
18.
Neuropsychopharmacology ; 33(7): 1584-602, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17805310

ABSTRACT

Chromatin remodeling by posttranslational modification of histones plays an important role in brain plasticity, including memory, response to stress and depression. The importance of H3/4 histones acetylation by CREB-binding protein (CBP) or related histone acetyltransferase, including p300, was specifically demonstrated using knockout (KO) mouse models. The physiological role of a related protein that also acts as a transcriptional coactivator with intrinsic histone acetylase activity, the p300/CBP-associated factor (PCAF), is poorly documented. We analyzed the behavioral phenotype of homozygous male and female PCAF KO mice and report a marked impact of PCAF deletion on memory processes and stress response. PCAF KO animals showed short-term memory deficits at 2 months of age, measured using spontaneous alternation, object recognition, or acquisition of a daily changing platform position in the water maze. Acquisition of a fixed platform location was delayed, but preserved, and no passive avoidance deficit was noted. No gender-related difference was observed. These deficits were associated with hippocampal alterations in pyramidal cell layer organization, basal levels of Fos immunoreactivity, and MAP kinase activation. PCAF KO mice also showed an exaggerated response to acute stress, forced swimming, and conditioned fear, associated with increased plasma corticosterone levels. Moreover, learning and memory impairments worsened at 6 and 12 months of age, when animals failed to acquire the fixed platform location in the water maze and showed passive avoidance deficits. These observations demonstrate that PCAF histone acetylase is involved lifelong in the chromatin remodeling necessary for memory formation and response to stress.


Subject(s)
Memory/physiology , Stress, Psychological/genetics , Stress, Psychological/physiopathology , p300-CBP Transcription Factors/deficiency , Age Factors , Analysis of Variance , Animals , Avoidance Learning/physiology , Behavior, Animal , Conditioning, Psychological/physiology , Corticosterone/blood , Disease Models, Animal , Emotions/physiology , Exploratory Behavior/physiology , Fear , Female , Hippocampus/pathology , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Knockout , Pattern Recognition, Visual/physiology , Sex Factors , Stress, Psychological/pathology
19.
Clin Immunol ; 119(1): 38-50, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16426893

ABSTRACT

A baculovirus-expressed chimeric recombinant IgG1 (rIgG1) antibody, with Cgamma1 and Ckappa human constant domains, was derived from the murine monoclonal antibody (mAb) 13B8.2, which is specific for the CDR3-like loop of the CD4 molecule and which inhibits HIV-1 replication. Chimeric rIgG1 antibody 13B8.2 blocked, in a dose-dependent manner, antigen presentation through inhibition of subsequent IL-2 secretion by stimulated T cells. The one-way mixed lymphocyte reaction was abrogated by previous addition of baculovirus-produced rIgG1 13B8.2 in the T-cell culture. Anti-proliferative activity of rIgG1 was demonstrated on CD3-activated CD4+ T lymphocytes from healthy donors, such effect being associated with reduced IL-2 secretion of activated T cells. On the other hand, no proliferation inhibition was observed on CD4+ T lymphocytes activated with phorbol ester plus ionomycin, suggesting that rIgG1 13B8.2 preferentially acts on a proximal TCR-induced signaling pathway. Treatment of DBA1/J human CD4-transgenic mice with 100 microg of recombinant antibody for three consecutive days led to in vivo recovery of rIgG1 antibody 13B8.2 both coated on murine T lymphocytes and free in mouse serum, without CD4 depletion or down-modulation. These findings predict that the baculovirus-expressed chimeric rIgG1 anti-CD4 antibody 13B8.2 is a promising candidate for immunotherapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD4 Antigens/immunology , Immunoglobulin G/immunology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/drug effects , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Formation/immunology , Antigen Presentation/drug effects , Antigen-Antibody Reactions/immunology , Baculoviridae/genetics , CD3 Complex/immunology , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation/drug effects , Epitopes/genetics , Epitopes/immunology , Gene Expression/drug effects , HIV Long Terminal Repeat/genetics , HeLa Cells , Humans , Immunization, Passive , Interleukin-2/metabolism , Ionomycin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred DBA , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
20.
Proc Natl Acad Sci U S A ; 102(38): 13586-91, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16174749

ABSTRACT

Patients with severe combined immunodeficiency (SCID) present with opportunistic infections that are almost universally fatal in infancy. The mainstay treatment for these patients is allogeneic hematopoietic stem cell (HSC) transplantation, but sustained polyclonal T cell reconstitution is too often unsatisfactory. Although transplantation is conventionally performed by i.v. administration of HSC, we hypothesized that an intrathymic strategy would be superior. Indeed, several progenitor cell populations are incapable of homing to the thymus, the major site of T cell differentiation, and it appears that there are extensive time periods during which the thymus is refractory to progenitor cell import. To test this hypothesis, nonconditioned infant ZAP-70-deficient SCID mice were intrathymically injected with WT bone marrow progenitor cells, a procedure accomplished without surgical intervention. Upon intrathymic HSC injection, there was a more rapid T cell differentiation, with mature thymocytes detected by 4 weeks after transplantation. Intrathymic injection of HSC also resulted in significantly higher numbers of peripheral T cells, increased percentages of naïve T cells, and more diverse T cell receptor repertoires. Moreover, T cell reconstitution after intrathymic transplantation was obtained after injection of 10-fold fewer donor HSC. Thus, this intrathymic transplantation approach may improve the outcome of SCID patients by enhancing T cell reconstitution.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Protein-Tyrosine Kinases/deficiency , Recovery of Function/immunology , Severe Combined Immunodeficiency/therapy , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Humans , Mice , Mice, Knockout , Severe Combined Immunodeficiency/immunology , Thymus Gland/immunology , Transplantation, Homologous , ZAP-70 Protein-Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...