Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Infect Dis Now ; 52(7): 414-417, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36089286

ABSTRACT

INTRODUCTION: Vancomycin prescription modalities remain non-consensual. We examined and evaluated the vancomycin prescription habits of infectious disease specialists in France. METHODS: Through an anonymized online questionnaire sent to members of the French Infectious Diseases Society, detailed information on vancomycin prescription modalities was collected. RESULTS: Out of the 712 physicians contacted, 179 (25%) completed the questionnaire; 174 (97%) of them routinely prescribed intravenous vancomycin: 95 (55%) by continuous infusion only, 12 (7%) by intermittent infusion, while 67 (38%) used the two modalities. Among continuous administration users, 157 (97%) applied a loading dose of 15 mg/kg or less (n = 80, 49%), 20-25 mg/kg (n = 33, 20%), or 30 mg/kg or more (n = 45, 28%); 143 (88%) used a maintenance dosage of 30 mg/kg/day and 157 (97%) carried out drug monitoring. CONCLUSION: In France, infectious disease specialists favor continuous administration of vancomycin using a loading dose, with systematic monitoring of vancomycin serum concentrations.


Subject(s)
Physicians , Vancomycin , Humans , Vancomycin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Drug Monitoring , Prescriptions
3.
Nat Med ; 28(1): 71-80, 2022 01.
Article in English | MEDLINE | ID: mdl-35075289

ABSTRACT

Patients with Wiskott-Aldrich syndrome (WAS) lacking a human leukocyte antigen-matched donor may benefit from gene therapy through the provision of gene-corrected, autologous hematopoietic stem/progenitor cells. Here, we present comprehensive, long-term follow-up results (median follow-up, 7.6 years) (phase I/II trial no. NCT02333760 ) for eight patients with WAS having undergone phase I/II lentiviral vector-based gene therapy trials (nos. NCT01347346 and NCT01347242 ), with a focus on thrombocytopenia and autoimmunity. Primary outcomes of the long-term study were to establish clinical and biological safety, efficacy and tolerability by evaluating the incidence and type of serious adverse events and clinical status and biological parameters including lentiviral genomic integration sites in different cell subpopulations from 3 years to 15 years after gene therapy. Secondary outcomes included monitoring the need for additional treatment and T cell repertoire diversity. An interim analysis shows that the study meets the primary outcome criteria tested given that the gene-corrected cells engrafted stably, and no serious treatment-associated adverse events occurred. Overall, severe infections and eczema resolved. Autoimmune disorders and bleeding episodes were significantly less frequent, despite only partial correction of the platelet compartment. The results suggest that lentiviral gene therapy provides sustained clinical benefits for patients with WAS.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cell Transplantation , Lentivirus/genetics , Wiskott-Aldrich Syndrome/therapy , Adolescent , Adult , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Infant , Treatment Outcome , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Young Adult
6.
Rev Med Interne ; 39(11): 863-868, 2018 Nov.
Article in French | MEDLINE | ID: mdl-29628173

ABSTRACT

Plague is a bacterial zoonosis caused by Yersinia pestis, usually found in fleas and small rodents that constitute the reservoir of the disease. It is transmitted to humans by flea bite, contact with rodents or inhalation of infected droplets. There are three clinical forms: bubonic plague, pulmonary plague and septicemic plague. The usual presentation is a flu-like syndrome possibly accompanied by an inflammatory lymphadenopathy which appears after 1 to 7days of incubation. Bubonic plague has a case fatality rate of about 50% while other forms of plague are almost always fatal without treatment. Diagnosis can be confirmed by usual bacteriological techniques (Gram examination, culture) but also by serological examination, use of rapid diagnostic tests or PCR. Although aminoglycosides are traditionally regarded as the most effective treatment, fluoroquinolones or cyclins are currently recommended in France. Plague is one of the re-emerging diseases according to the WHO and Madagascar suffered in 2017 the most important plague epidemic of the 21st century with more than 2000 cases and 200 deaths. Peru and the Democratic Republic of Congo are also considered endemic areas. Public health measures and a relentless fight against poverty are the cornerstone of the control of the disease. Vaccine improvement in endemic areas may also play an important role.


Subject(s)
Plague , Animals , Anti-Bacterial Agents/therapeutic use , France/epidemiology , Humans , Plague/diagnosis , Plague/epidemiology , Plague/microbiology , Plague/therapy , Siphonaptera/microbiology , Yersinia pestis/genetics , Yersinia pestis/isolation & purification
7.
Gene Ther ; 23(2): 176-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26361272

ABSTRACT

Artemis is a factor of the non-homologous end joining pathway involved in DNA double-strand break repair that has a critical role in V(D)J recombination. Mutations in DCLRE1C/ARTEMIS gene result in radiosensitive severe combined immunodeficiency in humans owing to a lack of mature T and B cells. Given the known drawbacks of allogeneic hematopoietic stem cell transplantation (HSCT), gene therapy appears as a promising alternative for these patients. However, the safety of an unregulated expression of Artemis has to be established. We developed a transgenic mouse model expressing human Artemis under the control of the strong CMV early enhancer/chicken beta actin promoter through knock-in at the ROSA26 locus to analyze this issue. Transgenic mice present a normal development, maturation and function of T and B cells with no signs of lymphopoietic malignancies for up to 15 months. These results suggest that the over-expression of Artemis in mice (up to 40 times) has no deleterious effects in early and mature lymphoid cells and support the safety of gene therapy as a possible curative treatment for Artemis-deficient patients.


Subject(s)
Endonucleases/genetics , Lymphopoiesis , T-Lymphocytes/cytology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins , Endonucleases/therapeutic use , Genetic Therapy , Humans , Immunoglobulin Class Switching/genetics , Lymphopoiesis/genetics , Mice , Mice, Transgenic , Severe Combined Immunodeficiency/therapy , T-Lymphocytes/immunology
8.
Gene Ther ; 21(9): 828-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25030611

ABSTRACT

Recombinant adeno-associated viral vectors (rAAVs) are used for therapeutic gene transfer in skeletal muscle, but it is unclear if immune reactivity to gene transfer and persistence of transgene are affected by pathologic conditions such as muscular dystrophy. Thus, we compared dystrophic mice devoid of α-sarcoglycan with healthy mice to characterize immune cell activation and cellular populations contributing to the loss of gene-modified myofibers. Following rAAV2/1 delivery of an immunogenic α-sarcoglycan reporter transgene in the muscle, both strains developed strong CD4 and CD8 T-cell-mediated immune responses in lymphoid organs associated with muscle CD3+ T and CD11b+ mononuclear cell infiltrates. Selective cell subset depletion models revealed that CD4+ T cells were essential for transgene rejection in both healthy and pathologic mice, but macrophages and CD8+ T cells additionally contributed as effector cells of transgene rejection only in dystrophic mice. Vectors restricting transgene expression in antigen-presenting cells showed that endogenous presentation of transgene products was the sole mechanism responsible for T-cell priming in normal mice, whereas additional and protracted antigenic presentation occurred in dystrophic animals, leading to secondary CD4+ T-cell activation and failure to maintain transgene expression. Therefore, the dystrophic environment diversifies cellular immune response mechanisms induced by gene transfer, with a negative outcome.


Subject(s)
Dependovirus/genetics , MicroRNAs/genetics , Muscles/immunology , Muscular Dystrophy, Animal/immunology , Sarcoglycans/genetics , Animals , Antigen-Presenting Cells/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dependovirus/immunology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Muscles/pathology , Muscular Dystrophy, Animal/therapy , Sarcoglycans/metabolism
9.
Gene Ther ; 18(11): 1087-97, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21544095

ABSTRACT

Insertional mutagenesis represents a serious adverse effect of gene therapy with integrating vectors. However, although uncontrolled activation of growth-promoting genes in stem cells can predictably lead to oncological processes, this is far less likely if vector transcriptional activity can be restricted to fully differentiated cells. Diseases requiring phenotypic correction only in mature cells offer such an opportunity, provided that lineage/stage-restricted systems can be properly tailored. In this study, we followed this reasoning to design lentiviral vectors for the gene therapy of chronic granulomatous disease (CGD), an immune deficiency due a loss of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in phagocytes, most often secondary to mutations in gp91(phox). Using self-inactivating HIV1-derived vectors as background, we first expressed enhanced green fluorescent protein (eGFP) from a minimal gp91(phox) promoter, adding various natural or synthetic transcriptional regulatory elements to foster both specificity and potency. The resulting vectors were assessed either by transplantation or by lentiviral transgenesis, searching for combinations conferring strong and specific expression into mature phagocytic cells. The most promising vector was modified to express gp91(phox) and used to treat CGD mice. High-level restoration of NADPH activity was documented in granulocytes from the treated animals. We propose that this lineage-specific lentiviral vector is a suitable candidate for the gene therapy of CGD.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Granulomatous Disease, Chronic/therapy , Lentivirus/genetics , Receptors, Immunologic/genetics , Animals , Gene Transfer Techniques , Granulocytes/metabolism , Mice , NADPH Oxidases/metabolism
10.
Gene Ther ; 18(5): 479-87, 2011 May.
Article in English | MEDLINE | ID: mdl-21160533

ABSTRACT

Lentiviral vectors are effective tools for gene transfer and integrate variable numbers of proviral DNA copies in variable proportions of cells. The levels of transduction of a cellular population may therefore depend upon experimental parameters affecting the frequency and/or the distribution of vector integration events in this population. Such analysis would require measuring vector copy numbers (VCN) in individual cells. To evaluate the transduction of hematopoietic progenitor cells at the single-cell level, we measured VCN in individual colony-forming cell (CFC) units, using an adapted quantitative PCR (Q-PCR) method. The feasibility, reproducibility and sensitivity of this approach were tested with characterized cell lines carrying known numbers of vector integration. The method was validated by correlating data in CFC with gene expression or with calculated values, and was found to slightly underestimate VCN. In spite of this, such Q-PCR on CFC was useful to compare transduction levels with different infection protocols and different vectors. Increasing the vector concentration and re-iterating the infection were two different strategies that improved transduction by increasing the frequency of transduced progenitor cells. Repeated infection also augmented the number of integrated copies and the magnitude of this effect seemed to depend on the vector preparation. Thus, the distribution of VCN in hematopoietic colonies may depend upon experimental conditions including features of vectors. This should be carefully evaluated in the context of ex vivo hematopoietic gene therapy studies.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cells , Lentivirus/genetics , Antigens, CD34/metabolism , Cell Line , Gene Transfer Techniques , Humans , Stem Cells , Transduction, Genetic , Virus Integration
11.
Gene Ther ; 16(5): 605-19, 2009 May.
Article in English | MEDLINE | ID: mdl-19262615

ABSTRACT

The woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) is widely used in retroviral gene transfer vectors. However, this element contains an open-reading frame (ORF) encoding a truncated peptide of the woodchuck hepatitis virus X protein (WHX). Because we are developing a lentiviral vector for the gene therapy of Wiskott-Aldrich syndrome (WAS), we evaluated whether the WPRE was needed in the gene transfer cassette and tested the possibility of replacing it with a mutated derivative. The transcriptional activity of the WPRE was undetectable in the context of the lentiviral vector but the element was capable of translating a polypeptide. This capability was abrogated by mutating the WHX ORF translation start. The WPRE was required to express high levels of the transgene and for that, the native form or mutated derivatives functioned equivalently. The vector using a WAS gene promoter and the mut6 WPRE induced long-term expression of the WAS transgene in vivo, correcting cytoskeletal defects, thymocyte and B-cell numbers and improved the colitis of WAS-null mice. By providing additional evidence of efficacy of this WAS lentiviral vector with improved safety features, our results validate a mutated WPRE, which should be useful in future gene therapy applications.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hepatitis B Virus, Woodchuck/genetics , Trans-Activators/genetics , Wiskott-Aldrich Syndrome/therapy , Animals , Cell Line , Colitis/pathology , Colitis/therapy , Gene Expression Regulation , Humans , Lentivirus/genetics , Mice , Mutation , Open Reading Frames/genetics , Plasmids/genetics , Trans-Activators/biosynthesis , Transduction, Genetic , Transgenes , Wiskott-Aldrich Syndrome/pathology
12.
Gene Ther ; 14(5): 415-28, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17051251

ABSTRACT

Gene therapy has been proposed as a potential treatment for Wiskott-Aldrich syndrome (WAS), a severe primary immune deficiency characterized by multiple hematopoietic-specific cellular defects. In order to develop an optimal lentiviral gene transfer cassette for this application, we compared the performance of several internal promoters in a variety of cell lineages from human WAS patients. Vectors using endogenous promoters derived from short (0.5 kb) or long (1.6 kb) 5' flanking sequences of the WAS gene, expressed the transgene in T, B, dendritic cells as well as CD34(+) progenitor cells, but functioned poorly in non-hematopoietic cells. Defects of T-cell proliferation and interleukin-2 production, and the cytoskeletal anomalies in WAS dendritic cells were also corrected. The levels of reconstitution were comparable to those obtained following transduction with similar lentiviral vectors incorporating constitutive PGK-1, EF1-alpha promoters or the spleen focus forming virus gammaretroviral LTR. Thus, native regulatory sequences target the expression of the therapeutic WAS transgene to the hematopoietic system, as is naturally the case for WAS, and are effective for correction of multiple cellular defects. These vectors may have significant advantages for clinical application in terms of natural gene regulation, and reduction in the potential for adverse mutagenic events.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic/methods , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/therapy , Antigens, CD34/immunology , B-Lymphocytes/metabolism , Base Sequence , Blotting, Western/methods , Cell Line , Cell Proliferation , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression , Gene Targeting/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hematopoietic Stem Cells/immunology , Humans , Interleukin-2/immunology , Microscopy, Fluorescence , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein/analysis , Wiskott-Aldrich Syndrome Protein/genetics
13.
Gene Ther ; 12(7): 597-606, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15616597

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is an immune deficiency with thrombopenia resulting from mutations in the WASP gene. This gene normally encodes the Wiskott-Aldrich syndrome protein (WASP), a major cytoskeletal regulator expressed in hematopoietic cells. Gene therapy is a promising option for the treatment of WAS, requiring that clinically applicable WASP gene transfer vectors demonstrate efficacy in preclinical studies. Here, we describe a self-inactivating HIV-1-derived lentiviral vector encoding human WASP and show that it effectively transduced bone marrow progenitor cells of WASP knockout (WKO) mice. Transplantation of these transduced cells into lethally irradiated WKO recipients led to stable expression of WASP and correction of immune, inflammatory and cytoskeletal defects. Splenic T-cell proliferation was restored, podosomes were reinstated on bone-marrow-derived dendritic cells and colon inflammation was reduced. This shows for the first time (a) that cytoskeletal defects can be corrected in WKO mice, (b) that human WASP is biologically active in mice and (c) that a lentiviral vector is effective to express human WASP in vivo over several months. These data support further development of such lentiviral vectors for the gene therapy of WAS.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/therapeutic use , HIV-1/genetics , Proteins/genetics , Wiskott-Aldrich Syndrome/therapy , Animals , Bone Marrow Transplantation/methods , Colitis/therapy , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Gene Expression , Genetic Therapy/adverse effects , Humans , Mice , Mice, Knockout , Proteins/metabolism , RNA, Messenger/genetics , T-Lymphocytes/immunology , Transduction, Genetic , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein
14.
Int Immunol ; 13(8): 1053-61, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11470775

ABSTRACT

The cytokine IL-12, a product of dendritic cells (DC), plays a major role in cellular immunity, notably by inducing lymphocytes to produce IFN-gamma. Microbial products, T cell signals and cytokines induce the production of IL-12. Here, IL-1 beta is identified as a new IL-12-inducing agent, acting conjointly with CD40 ligand (CD40L) on human monocyte-derived DC in vitro. The effects of IL-1 beta were dose dependent, specifically blocked by neutralizing antibodies, and were observed both in immature and mature DC. Immature DC secreted more IL-12 than mature DC, but the effects of IL-1 beta were not due to a block of DC maturation as determined by analysis of DC surface markers. The mechanisms of action of IL-1 beta could be contrasted to that of other inducers of IL-12 such as IFN-gamma and lipopolysaccharide (LPS). Either IL-1 beta or IFN-gamma co-induced IL-12 with CD40L but conjointly, IL-1 beta, CD40L and IFN-gamma synergized, inducing very high levels of IL-12. The effects of IL-1 beta differed from those of LPS in that IL-1 beta, unlike LPS, could not induce IL-12 solely after IFN-gamma priming; and when combined with CD40L, IL-1 beta, unlike LPS, induced little IL-10. The mechanism of action of IL-1 beta involves IL-12 alpha mRNA up-regulation, and we show that the combination of CD40L and IL-1 beta induces high levels of IL-12 alpha and IL-12 beta mRNA in DC. Altogether, these results delineate a new mechanism linking adaptive and innate immune responses for the regulation of IL-12 production in DC and for the role of IL-1 beta in the development of cellular immunity.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Interleukin-12/biosynthesis , Interleukin-1/pharmacology , Antibodies, Blocking , Antibody Specificity , CD40 Ligand/pharmacology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dimerization , Dose-Response Relationship, Immunologic , Gene Expression Regulation/immunology , Humans , Immune Sera/pharmacology , Interferon-gamma/immunology , Interleukin-1/antagonists & inhibitors , Interleukin-1/immunology , Interleukin-12/antagonists & inhibitors , Interleukin-12/genetics , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology
15.
Mech Dev ; 104(1-2): 105-11, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11404085

ABSTRACT

We cloned a novel murine gene, designated Hemogen (hemopoietic gene), which was sequentially expressed in active hematopoietic sites and downregulated in the process of blood cell differentiation. Hemogen transcripts were specifically detected in blood islands, primitive blood cells and fetal liver during embryogenesis, and then remained in bone marrow and spleen in adult mice. Immunostaining demonstrated that Hemogen was a nuclear protein. We also identified a human homologue of Hemogen, named EDAG, which was mapped to chromosome 9q22, a leukemia breakpoint. Like Hemogen, EDAG exhibited specific expression in hematopoietic tissues and cells. Taken together, these data are consistent with Hemogen and EDAG playing an important role in hematopoietic development and neoplasms.


Subject(s)
Cell Nucleus/metabolism , Chromosomes, Human, Pair 9 , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cells/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Bone Marrow/metabolism , COS Cells , Cloning, Molecular , Down-Regulation , Humans , In Situ Hybridization , Liver/embryology , Mice , Molecular Sequence Data , Plasmids , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spleen/metabolism , Tissue Distribution
16.
Cell Immunol ; 208(2): 115-24, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11333144

ABSTRACT

When dendritic cells (DC) present antigens to T cells, reciprocal cellular interactions occur that lead to cytokine production. This cytokine response is regulated by specific properties of DC, notably their maturation/activation status and perhaps their origin. The latter possibility prompted us to determine if DC produced along distinct developmental pathways induced distinct T cell responses. Hematopoietic progenitor cells with the potential to differentiate into multiple lineages of cells were induced to differentiate into DC along two pathways. One leads to the formation of lymphoid-related DC but not of monocyte-derived DC and is induced by culture of CD34(+) cells with flt-3 ligand (F), c-kit ligand (K), GM-CSF (Gm), IL-1beta ("1"), and IL-7 ("7") (FKGm17). Another pathway with distinct molecular requirements supports in part monocyte-derived DC and is induced by the cytokines F, K, Gm, TNF-alpha (T), and IL-4 ("4") (FKGmT4). DC produced along these two pathways were isolated by flow cytometry and compared. They differed only slightly in phenotype and morphology and both induced Th1-type cytokine production in MLR (mixed lymphocyte reactions). However, on a cell-per-cell basis, FKGm17-DC produced more IL-18 or IL-12 and induced more IFN-gamma by T cells in MLR. Such superior properties were not intrinsically determined by the origin of the DC but were induced by FKGm17 cytokines. We conclude that lymphoid-related DC have the potential to induce Th1 T cell responses but that environmental signals strongly influence T-cell-stimulating properties of DC.


Subject(s)
Dendritic Cells/immunology , Hematopoietic Stem Cells/immunology , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-18/biosynthesis , T-Lymphocytes/immunology , Adult , Antigens, CD34/immunology , Biomarkers , Cell Membrane/immunology , Cytokines/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-1/immunology , Interleukin-7/immunology , Membrane Proteins/immunology , Stem Cell Factor/immunology
17.
J Leukoc Biol ; 69(4): 675-83, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11310856

ABSTRACT

Ikaros family members play critical roles in hematopoietic development, yet molecules regulated by Ikaros proteins remain incompletely characterized. To determine the requirements for functional Ikaros proteins, we overexpressed Ik7, a dominant negative Ikaros protein, in human cell lines and hematopoietic progenitor cells. Ik7 is known to block the normal function of other Ikaros family members in human and mouse cells. Retroviral-mediated overexpression of Ik7 affected two distinct, migratory properties of the CEM T-cell line. Ik7 down-regulated L-selectin cell-surface expression, an effect not a result of increased shedding but of a decrease in L-selectin mRNA levels. Ik7 also reduced the spontaneous migration of CEM T cells in 3-D collagen gels. A reduction in L-selectin, cell-surface expression was also induced by Ik7 in CD34+ hematopoietic progenitor cells. In contrast, the Reh B cell line showed an up-regulation of L-selectin, cell-surface levels when expressing Ik7. For the first time, this study defines an effect of Ikaros proteins in the control of migration-related properties and shows that intact Ikaros proteins are important in a cell type-specific manner for the normal regulation of L-selectin expression.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/metabolism , L-Selectin/biosynthesis , T-Lymphocytes/metabolism , Transcription Factors/physiology , Animals , Antigens, CD/biosynthesis , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line , Cell Movement , Collagen , Down-Regulation , Exons/genetics , Gels , Gene Expression Regulation, Leukemic/physiology , Genes, Dominant , Genes, Reporter , Hematopoiesis/physiology , Humans , Ikaros Transcription Factor , L-Selectin/genetics , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , T-Lymphocytes/cytology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Cells, Cultured
18.
Talanta ; 54(2): 307-17, 2001 Apr 12.
Article in English | MEDLINE | ID: mdl-18968253

ABSTRACT

Different mass analysers [(quadrupole (Q), time-of-flight (TOF) and multicollector (MC) sector-field (SF)] of ions produced in an inductively coupled plasma were evaluated for the determination of lead isotope ratios in wine samples. A population of 20 wines of different origin including two reference wines from the EC Standards, Measurement and Testing Programme with concentrations varying between 7-140 mug Pb l(-1) was investigated. Wines were analyzed directly by Q ICP MS and MC ICP MS. The poor sensitivity of the TOF instrument, further aggravated by matrix signal suppression, did not allow the acquisition of data for wine samples that contained less than 50 mug l(-1) in the direct sample introduction mode. The separation and preconcentration of lead were therefore required. The precision obtained for the (206)Pb/(207)Pb and (208)Pb/(206)Pb were similar and equal to 0.14-2.7% for Q ICP MS, 0.04-0.17% for TOF ICP MS and 0.01-0.12% for MC ICP MS. The precision for (206)Pb/(204)Pb was 0.44-5.29, 0.15-1.7, 0.08-1.6%, respectively. On the level of accuracy, the data from TOF ICP MS and MC ICP MS were in good agreement. The accuracy of Q ICP MS data was judged satisfactory in comparison with the other techniques but their poor precision was a significant obstacle on the way of using these data for the determination of the geographic origin of wine.

19.
Methods Mol Med ; 64: 275-81, 2001.
Article in English | MEDLINE | ID: mdl-21374267

ABSTRACT

Dendritic cells (DC) are "professional" antigen presenting cells (APC) that are pivotal for the initiation of T-cell-dependent immune responses (1). DC are widely distributed cells in the body, but are scarce and thus difficult to purify. Methods have become available to grow DC in culture, facilitating the study of their biological and developmental properties. One such method is described in this chapter to generate DC in vitro from CD34(+) Lineage(-) (Lin(-)) CD10(+) bone marrow (BM) progenitor cells that are easily committed to the lymphoid lineages. This source of DC precursors thus distinguishes itself from mature monocytes or from total CD34(+) cells, which have also been employed to generate DC in vitro (2-5). It is unclear at this point if DC produced from lymphoid progenitor cells or from monocytes or total CD34(+) cells are equivalent. Lymphoid progenitor cells displaying the cell-surface phenotype CD34(+) Lin(-) CD10(+) rapidly lose CD34 cell-surface expression in culture and quickly differentiate into T-, B-, or natural-killer (NK) lymphocytes in the appropriate SCID-hu assay or culture systems (6,7; Galy, unpublished observations). This population of CD34(+) BM progenitor cells contains cells expressing the IL-7Rα chain, TdT, RAG-1, the transcription factors Pax5 and Ikaros, and represent the earliest recognizable human B-cell precursor identified in human BM (7,8). BM cells of CD34(+) Lin(-) CD10(+) phenotype can differentiate into antigen-presenting DC under the appropriate culture conditions, generating "lymphoidrelated DC" (6). Limiting dilution analyses demonstrated that single clones of CD34(+) Lin(-) CD10(+) progenitor cells can generate B cells, NK cells, and DC suggesting that these three lineages of cells share a common developmental stage in the BM and thus have close developmental relationships (6). In spite of being multipotent cells, CD34(+) Lin- CD10(+) cells do not differentiate into myeloid or erythroid cells in the presence of cytokines permissive for the production of such lineages from CD34(+) cells (6,7). In contrast, cytokine combinations such as c-kit-ligand+GM-CSF+flt3-ligand+IL-1+IL-7, which cause total CD34(+) cells to become myeloid cells, induce CD34(+) Lin(-) CD10(+)to differentiate into DC. Such lymphoid-related DC display characteristic long membrane processes, have immunostimulatory properties, and express on the cell surface high levels of MHC class II molecules, CD 1a, CD40 with little or no CD14, CD19, or CD15 antigens. Owing to their distinct hematopoietic origin, lymphoid-related DC may represent a separate population of DC. It is tempting to speculate that lymphoid-related DC may develop in lymphoid organs such as the thymus where progenitor cells with similar dual lymphoid/DC potential have been described (9). The method described here consists of preparing mononuclear cells (MNC) from BM that are depleted by negative selection of cells expressing lineage markers (CD2, CD3, CD19, CD20, CD14, CD15, CD32, glycophorin A), found on T cells, B cells, monocytes, and erythrocytes. Lineage-depleted cells are labeled with fluorochrome-conjugated antibodies and CD34(+) Lin(-) CD10(+)cells are purified by flow cytometry. Sorted cells are placed in culture in the presence of Flt3-ligand, c-kit-ligand, GM-CSF, IL-1, and IL-7 for 2 wk to generate lymphoid-related DC.

20.
Genomics ; 68(1): 13-21, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10950922

ABSTRACT

Phemx (Pan hematopoietic expression) is a novel murine gene expressed in developmentally regulated sites of hematopoiesis from early in embryogenesis through adulthood. Phemx is expressed in hematopoietic progenitors and mature cells of the three main hematopoietic lineages. Conceptual translation of the murine Phemx cDNA predicts a 25-kDa polypeptide with four hydrophobic regions and several potential phosphorylation sites, suggestive of a transmembrane protein involved in cell signaling. The PHEMX protein is structurally similar to tetraspanin CD81 (TAPA-1), a transmembrane protein involved in leukocyte activation, adhesion, and proliferation. Phemx maps to the distal region of chromosome 7, a segment of the mouse genome that contains a cluster of genes that exhibit genomic imprinting. However, imprinting analysis of Phemx at the whole organ level shows that it is biallelically expressed, suggesting that mechanisms leading to monoallelic expression are not imposed at this locus. The human PHEMX ortholog is specifically expressed in hematopoietic organs and tissues and, in contrast to murine Phemx, undergoes alternative splicing. The unique mode and range of Phemx expression suggest that it plays a role in hematopoietic cell function.


Subject(s)
Chromosomes/genetics , Genomic Imprinting , Hematopoietic Stem Cells/metabolism , Membrane Proteins/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization , Jurkat Cells , K562 Cells , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muridae , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tetraspanins , Tissue Distribution , Tumor Cells, Cultured , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...