Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 19(9): 973-985, 2018 09.
Article in English | MEDLINE | ID: mdl-30127434

ABSTRACT

Human inborn errors of IFN-γ immunity underlie mycobacterial diseases. We describe patients with Mycobacterium bovis (BCG) disease who are homozygous for loss-of-function mutations of SPPL2A. This gene encodes a transmembrane protease that degrades the N-terminal fragment (NTF) of CD74 (HLA invariant chain) in antigen-presenting cells. The CD74 NTF therefore accumulates in the HLA class II+ myeloid and lymphoid cells of SPPL2a-deficient patients. This toxic fragment selectively depletes IL-12- and IL-23-producing CD1c+ conventional dendritic cells (cDC2s) and their circulating progenitors. Moreover, SPPL2a-deficient memory TH1* cells selectively fail to produce IFN-γ when stimulated with mycobacterial antigens in vitro. Finally, Sppl2a-/- mice lack cDC2s, have CD4+ T cells that produce small amounts of IFN-γ after BCG infection, and are highly susceptible to infection with BCG or Mycobacterium tuberculosis. These findings suggest that inherited SPPL2a deficiency in humans underlies mycobacterial disease by decreasing the numbers of cDC2s and impairing IFN-γ production by mycobacterium-specific memory TH1* cells.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Dendritic Cells/immunology , Membrane Proteins/metabolism , Mycobacterium Infections/immunology , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/physiology , Th1 Cells/immunology , Tuberculosis/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Cells, Cultured , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunity , Immunologic Memory , Infant , Interferon-gamma/metabolism , Lymphadenopathy , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mycobacterium Infections/genetics , Vaccination
2.
Nature ; 606(7913): 368-374, 2022 06.
Article in English | MEDLINE | ID: mdl-35418681

ABSTRACT

HIV-1 infection remains a public health problem with no cure. Anti-retroviral therapy (ART) is effective but requires lifelong drug administration owing to a stable reservoir of latent proviruses integrated into the genome of CD4+ T cells1. Immunotherapy with anti-HIV-1 antibodies has the potential to suppress infection and increase the rate of clearance of infected cells2,3. Here we report on a clinical study in which people living with HIV received seven doses of a combination of two broadly neutralizing antibodies over 20 weeks in the presence or absence of ART. Without pre-screening for antibody sensitivity, 76% (13 out of 17) of the volunteers maintained virologic suppression for at least 20 weeks off ART. Post hoc sensitivity analyses were not predictive of the time to viral rebound. Individuals in whom virus remained suppressed for more than 20 weeks showed rebound viraemia after one of the antibodies reached serum concentrations below 10 µg ml-1. Two of the individuals who received all seven antibody doses maintained suppression after one year. Reservoir analysis performed after six months of antibody therapy revealed changes in the size and composition of the intact proviral reservoir. By contrast, there was no measurable decrease in the defective reservoir in the same individuals. These data suggest that antibody administration affects the HIV-1 reservoir, but additional larger and longer studies will be required to define the precise effect of antibody immunotherapy on the reservoir.


Subject(s)
Anti-Retroviral Agents , HIV Antibodies , HIV Infections , HIV-1 , Viral Load , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/virology , HIV Antibodies/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/growth & development , Humans , Proviruses/drug effects , Viral Load/drug effects , Viremia/drug therapy , Virus Latency/drug effects
3.
Nature ; 591(7851): 639-644, 2021 03.
Article in English | MEDLINE | ID: mdl-33461210

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected 78 million individuals and is responsible for over 1.7 million deaths to date. Infection is associated with the development of variable levels of antibodies with neutralizing activity, which can protect against infection in animal models1,2. Antibody levels decrease with time, but, to our knowledge, the nature and quality of the memory B cells that would be required to produce antibodies upon reinfection has not been examined. Here we report on the humoral memory response in a cohort of 87 individuals assessed at 1.3 and 6.2 months after infection with SARS-CoV-2. We find that titres of IgM and IgG antibodies against the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 decrease significantly over this time period, with IgA being less affected. Concurrently, neutralizing activity in plasma decreases by fivefold in pseudotype virus assays. By contrast, the number of RBD-specific memory B cells remains unchanged at 6.2 months after infection. Memory B cells display clonal turnover after 6.2 months, and the antibodies that they express have greater somatic hypermutation, resistance to RBD mutations and increased potency, indicative of continued evolution of the humoral response. Immunofluorescence and PCR analyses of intestinal biopsies obtained from asymptomatic individuals at 4 months after the onset of coronavirus disease 2019 (COVID-19) revealed the persistence of SARS-CoV-2 nucleic acids and immunoreactivity in the small bowel of 7 out of 14 individuals. We conclude that the memory B cell response to SARS-CoV-2 evolves between 1.3 and 6.2 months after infection in a manner that is consistent with antigen persistence.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunity, Humoral/immunology , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Biopsy , COVID-19/blood , Cohort Studies , Fluorescent Antibody Technique , Humans , Immunity, Humoral/genetics , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunologic Memory/immunology , Intestines/immunology , Middle Aged , Mutation , Somatic Hypermutation, Immunoglobulin , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Time Factors , Young Adult
4.
Blood ; 121(25): 5034-44, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23482932

ABSTRACT

Functional differences between human dendritic cell (DC) subsets and the potential benefits of targeting them with vaccines remain poorly defined. Here we describe that mice with reconstituted human immune system components (huNSG mice) develop all human conventional and plasmacytoid DC compartments in lymphoid organs. Testing different Toll-like receptor agonists for DC maturation in vivo, we found that IL-12p70 and interferon (IFN)-α production correlated with the maturation of CD141+ (BDCA3+) conventional DCs in huNSG mice. Furthermore, depletion of CD141+ DCs before stimulation significantly reduced IFN-α levels in vivo. This DC subset produced similar total amounts but different subtypes of IFN-α in response to synthetic double-stranded RNA compared with plasmacytoid DCs in response to a single-stranded RNA equivalent. Moreover, synthetic double-stranded RNA as adjuvant and antigen targeting to the endocytic receptor DEC-205, a combination that focuses antigen presentation for T-cell priming on CD141+ DCs, stimulated antigen-specific human CD4+ T-cell responses. Thus, the human CD141+ DC subset is a prominent source of IFN-α and interleukin-12 production and should be further evaluated for vaccine development.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Interferon-alpha/biosynthesis , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , RNA, Double-Stranded/immunology , Receptors, Cell Surface/immunology , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interferon-alpha/immunology , Mice , Minor Histocompatibility Antigens
5.
Nat Chem Biol ; 9(4): 250-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416331

ABSTRACT

Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient production of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.


Subject(s)
Adaptive Immunity/drug effects , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens/immunology , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Immunoconjugates/immunology , Lectins, C-Type/immunology , Poly dA-dT/immunology , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antigens/administration & dosage , Antigens/chemistry , Antigens, CD/administration & dosage , Antigens, CD/chemistry , Dendritic Cells/immunology , Drug Delivery Systems , Genetic Vectors , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Plasmids , Poly dA-dT/administration & dosage , Poly dA-dT/chemistry , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/chemistry
6.
J Immunol ; 191(5): 2194-204, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918986

ABSTRACT

Chronic activation of T cells is a hallmark of HIV-1 infection and plays an important role in disease progression. We previously showed that the engagement of the inhibitory receptor programmed death (PD)-1 on HIV-1-specific CD4(+) and CD8(+) T cells leads to their functional exhaustion in vitro. However, little is known about the impact of PD-1 expression on the turnover and maturation status of T cells during the course of the disease. In this study, we show that PD-1 is upregulated on all T cell subsets, including naive, central memory, and transitional memory T cells in HIV-1-infected subjects. PD-1 is expressed at similar levels on most CD4(+) T cells during the acute and the chronic phase of disease and identifies cells that have recently entered the cell cycle. In contrast, PD-1 expression is dramatically increased in CD8(+) T cells during the transition from acute to chronic infection, and this is associated with reduced levels of cell proliferation. The failure to downregulate expression of PD-1 in most T cells during chronic HIV-1 infection is associated with persistent alterations in the distribution of T cell subsets and is associated with impaired responses to IL-7. Our findings identify PD-1 as a marker for aberrant distribution of T cell subsets in HIV-1 infection.


Subject(s)
Biomarkers/analysis , HIV Infections/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocyte Subsets/immunology , Flow Cytometry , HIV Infections/metabolism , Humans , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocyte Subsets/metabolism
7.
Cell Rep ; 43(6): 114296, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823019

ABSTRACT

To explore the influence of genetics on homeostatic regulation of dendritic cell (DC) numbers, we present a screen of DCs and their progenitors in lymphoid and non-lymphoid tissues in Collaborative Cross (CC) and Diversity Outbred (DO) mice. We report 30 and 71 loci with logarithm of the odds (LOD) scores >8.18 and ranging from 6.67 to 8.19, respectively. The analysis reveals the highly polygenic and pleiotropic architecture of this complex trait, including many of the previously identified genetic regulators of DC development and maturation. Two SNPs in genes potentially underlying variation in DC homeostasis, a splice variant in Gramd4 (rs235532740) and a missense variant in Orai3 (rs216659754), are confirmed by gene editing using CRISPR-Cas9. Gramd4 is a central regulator of DC homeostasis that impacts the entire DC lineage, and Orai3 regulates cDC2 numbers in tissues. Overall, the data reveal a large number of candidate genes regulating DC homeostasis in vivo.


Subject(s)
Dendritic Cells , Quantitative Trait Loci , Animals , Dendritic Cells/metabolism , Mice , Quantitative Trait Loci/genetics , Polymorphism, Single Nucleotide , Mice, Inbred C57BL , Cell Count , Chromosome Mapping , Homeostasis
8.
Cell Rep ; 40(10): 111311, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36070690

ABSTRACT

Antiretroviral therapy controls, but does not cure, HIV-1 infection due to a reservoir of rare CD4+ T cells harboring latent proviruses. Little is known about the transcriptional program of latent cells. Here, we report a strategy to enrich clones of latent cells carrying intact, replication-competent HIV-1 proviruses from blood based on their expression of unique T cell receptors. Latent cell enrichment enabled single-cell transcriptomic analysis of 1,050 CD4+ T cells belonging to expanded clones harboring intact HIV-1 proviruses from 6 different individuals. The analysis reveals that most of these cells are T effector memory cells that are enriched for expression of HLA-DR, HLA-DP, CD74, CCL5, granzymes A and K, cystatin F, LYAR, and DUSP2. We conclude that expanded clones of latent cells carrying intact HIV-1 proviruses persist preferentially in a distinct CD4+ T cell population, opening possibilities for eradication.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , CD4-Positive T-Lymphocytes/metabolism , Clone Cells , DNA-Binding Proteins/metabolism , Gene Expression , HIV-1/genetics , HIV-1/metabolism , Humans , Nuclear Proteins/metabolism , Proviruses/genetics , Proviruses/metabolism , Virus Latency/genetics
9.
Nephrol Dial Transplant ; 26(9): 2852-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21292816

ABSTRACT

BACKGROUND: Glomerular filtration rate (GFR) decline with age increases the risk of inappropriate dosing of drugs. We investigated the determinants and the mortality associated with the use of drugs that are contraindicated or require dose adjustment according to kidney function among the community-dwelling elderly. METHODS: The Three-City population-based study included 8701 participants ≥65 years from 1999 to 2001. Exposure to the risk of inappropriate drug dosage was defined as reported use of either a contraindicated drug or one requiring dose adjustment according to the individual baseline glomerular filtration rate estimated (eGFR) with the Modification of Diet in Renal disease study equation. Six-year mortality was analysed using Cox models adjusted for several sociodemographic, biologic and clinical risk factors. RESULTS: The overall percentage of exposure to the risk of inappropriate drug use was 13.3% (contraindication, 0.8%): it was 52.5% (4.5%) in those with an eGFR of 30-59 and 96% (48%) in those <30 mL/min/1.73 m(2). Antihypertensive agents, fibrates and psycholeptics accounted for most of the drugs with dosing recommendations and antidiabetic agents and antihistamines for those contraindicated. Individuals at risk were more likely to be men, older, and under treatment for hypertension or hypercholesterolemia. Exposure to either risk was independently related to higher all-cause mortality (hazard ratio 1.4, 95% confidence interval 1.0-1.9) in participants with eGFR <60 mL/min/1.73 m(2). CONCLUSIONS: Contraindicated drug prescription was uncommon but >10% of the population took drugs requiring renal dosing adjustments. Regular monitoring of eGFR may prevent excess mortality associated with inappropriate drug prescription in the elderly.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Inappropriate Prescribing/adverse effects , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/pathology , Kidney/physiopathology , Aged , Cohort Studies , Community Health Planning , Female , Follow-Up Studies , France/epidemiology , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/epidemiology , Kidney Function Tests , Longitudinal Studies , Male , Prognosis , Prospective Studies , Risk Factors , Survival Rate
10.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33533915

ABSTRACT

SARS-CoV-2 is responsible for an ongoing pandemic that has affected millions of individuals around the globe. To gain further understanding of the immune response in recovered individuals, we measured T cell responses in paired samples obtained an average of 1.3 and 6.1 mo after infection from 41 individuals. The data indicate that recovered individuals show persistent polyfunctional SARS-CoV-2 antigen-specific memory that could contribute to rapid recall responses. Recovered individuals also show enduring alterations in relative overall numbers of CD4+ and CD8+ memory T cells, including expression of activation/exhaustion markers, and cell division.


Subject(s)
COVID-19/immunology , COVID-19/virology , Host-Pathogen Interactions/immunology , Immunity, Cellular , SARS-CoV-2/immunology , Adult , Aged , Antigens, Viral/immunology , Biomarkers , Female , Humans , Immunophenotyping , Lymphocyte Count , Male , Middle Aged , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
11.
bioRxiv ; 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33173867

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected 78 million individuals and is responsible for over 1.7 million deaths to date. Infection is associated with development of variable levels of antibodies with neutralizing activity that can protect against infection in animal models. Antibody levels decrease with time, but the nature and quality of the memory B cells that would be called upon to produce antibodies upon re-infection has not been examined. Here we report on the humoral memory response in a cohort of 87 individuals assessed at 1.3 and 6.2 months after infection. We find that IgM, and IgG anti-SARS-CoV-2 spike protein receptor binding domain (RBD) antibody titers decrease significantly with IgA being less affected. Concurrently, neutralizing activity in plasma decreases by five-fold in pseudotype virus assays. In contrast, the number of RBD-specific memory B cells is unchanged. Memory B cells display clonal turnover after 6.2 months, and the antibodies they express have greater somatic hypermutation, increased potency and resistance to RBD mutations, indicative of continued evolution of the humoral response. Analysis of intestinal biopsies obtained from asymptomatic individuals 4 months after coronavirus disease-2019 (COVID-19) onset, using immunofluorescence, or polymerase chain reaction, revealed persistence of SARS-CoV-2 nucleic acids and immunoreactivity in the small bowel of 7 out of 14 volunteers. We conclude that the memory B cell response to SARS-CoV-2 evolves between 1.3 and 6.2 months after infection in a manner that is consistent with antigen persistence.

12.
bioRxiv ; 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33330867

ABSTRACT

SARS-CoV-2 is responsible for an ongoing pandemic that affected millions of individuals around the globe. To gain further understanding of the immune response in recovered individuals we measured T cell responses in paired samples obtained an average of 1.3 and 6.1 months after infection from 41 individuals. The data indicate that recovered individuals show persistent polyfunctional SARS-CoV-2 antigen specific memory that could contribute to rapid recall responses. In addition, recovered individuals show enduring immune alterations in relative numbers of CD4 + and CD8 + T cells, expression of activation/exhaustion markers, and cell division. SUMMARY: We show that SARS-CoV-2 infection elicits broadly reactive and highly functional memory T cell responses that persist 6 months after infection. In addition, recovered individuals show enduring immune alterations in CD4 + and CD8 + T cells compartments.

13.
J Clin Immunol ; 29(5): 637-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19562472

ABSTRACT

INTRODUCTION: Due to their capacity to elicit and regulate immunity, dendritic cells (DCs) are important targets to improve vaccination. Knowing that programmed death-1 (PD-1) high virus-specific T cells become functionally exhausted during chronic exposure to human immunodeficiency virus-1 (HIV-1), the development of a therapeutic DC-based HIV-1 vaccine might include strategies that downregulate PD-L1 and PD-L2 counter-receptors. METHODS: After showing that monocyte-derived DCs rapidly upregulated PD-L1 and PD-L2 expression upon maturation with a variety of stimuli, e.g., Toll-like receptor ligands and cytokines, we determined that PD-L1 and PD-L2 expression could be knocked down by electroporation of a single small interfering RNA (siRNA) sequence twice at the monocyte and immature stages of DC development. This knockdown approached completion and was specific and lasting for several days. RESULTS: We then added the PD-L1 and PD-L2 silenced monocyte-derived DCs to peripheral blood mononuclear cells from HIV-1-infected individuals along with pools of 15-mer HIV-1 Gag p24 peptides. However, in cultures from six patients, there was only a modest enhancing effect of PD-L1 and PD-L2 silencing on CD8(+) T cell proliferative responses to the DCs. DISCUSSION: These findings suggest that, in monocyte-derived DCs, additional strategies than PD-L1 or PD-L2 blockade will be needed to improve the function of PD-1 high T cells.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/metabolism , HIV Infections/immunology , HIV-1/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , B7-H1 Antigen , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , HIV Core Protein p24/chemistry , HIV Core Protein p24/immunology , HIV Core Protein p24/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/pathogenicity , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/genetics , Monocytes/pathology , Monocytes/virology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Library , Programmed Cell Death 1 Ligand 2 Protein , RNA, Small Interfering/genetics
14.
J Am Med Inform Assoc ; 16(2): 203-10, 2009.
Article in English | MEDLINE | ID: mdl-19074305

ABSTRACT

OBJECTIVES: Medication errors constitute a major problem in all hospitals. Between 20% and 46% of prescriptions requiring dosage adjustments based on renal function are inappropriate. This study aimed to determine whether implementing alerts at the time of ordering medication integrated into the computerized physician order entry decreases the proportion of inappropriate prescriptions based on the renal function of inpatients. DESIGN: Six alternating 2-month control and intervention periods were conducted between August 2006 and August 2007 in two medical departments of a teaching hospital in France. A total of 603 patients and 38 physicians were included. During the intervention periods, alerts were triggered if a patient with renal impairment was prescribed one of the 24 targeted drugs that required adjustment according to estimated glomerular filtration rate (eGFR). MEASUREMENTS: The main outcome measure was the proportion of inappropriate first prescriptions, according to recommendation. RESULTS: A total of 1,122 alerts were triggered. The rate of inappropriate first prescriptions did not differ significantly between intervention and control periods (19.9% vs. 21.3%; p=0.63). The effect of intervention differed significantly between residents and senior physicians (p=0.03). Residents tended to make fewer errors in intervention versus control periods (Odds ratio 0.69; 95% confidence interval 0.41 to 1.15), whereas senior physicians tended to make more inappropriate prescriptions in intervention periods (odds ratio 1.88; 95% confidence interval 0.91 to 3.89). CONCLUSION: Alert activation was not followed by a significant decrease in inappropriate prescriptions in our study. Thus, it is still necessary to evaluate the impact of these systems if newly implemented in other settings thanks to studies also designed to watch for possible unanticipated effects of decision supports and their underlying causes.


Subject(s)
Drug Therapy, Computer-Assisted , Medical Order Entry Systems , Medication Errors/prevention & control , Pharmaceutical Preparations/administration & dosage , Reminder Systems , Renal Insufficiency/drug therapy , Drug Prescriptions , Female , France , Hospitals, Teaching/organization & administration , Humans , Male , Medication Systems, Hospital , Odds Ratio , Systems Integration
15.
Med Sci (Paris) ; 23(3): 279-84, 2007 Mar.
Article in French | MEDLINE | ID: mdl-17349289

ABSTRACT

Dendritic cells play a central role in the initiation of the immune response as they are the only antigen-presenting cells able to prime naive T cells. This makes the dendritic cells the vector of choice to use as a cell-based vaccine in immunotherapy. Although there are several strategies to deliver antigen to dendritic cells, the ones transfected with mRNA coding for tumor or viral antigens are able to induce potent antigen specific T-cell responses directed against multiple epitopes. In this review, we report several advances made in the field of anti-tumoral and anti-HIV immunotherapy using mRNA-transfected dendritic cells-based approaches.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Active , RNA, Messenger/genetics , Animals , Antigen Presentation , Antigens, CD/analysis , Antigens, Neoplasm/genetics , Antigens, Viral/genetics , Cells, Cultured/immunology , Cells, Cultured/metabolism , Clinical Trials, Phase I as Topic , Dendritic Cells/classification , Dendritic Cells/cytology , Dendritic Cells/metabolism , HIV Infections/therapy , Humans , Lymphocyte Activation , Mice , Myeloid Cells/cytology , Neoplasms/therapy , RNA, Neoplasm/genetics , RNA, Viral/genetics , T-Lymphocytes/immunology , Transfection
16.
J Exp Med ; 213(13): 2861-2870, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27864467

ABSTRACT

In humans, conventional dendritic cells (cDCs) exist as two unique populations characterized by expression of CD1c and CD141. cDCs arise from increasingly restricted but well-defined bone marrow progenitors that include the common DC progenitor that differentiates into the pre-cDC, which is the direct precursor of cDCs. In this study, we show that pre-cDCs in humans are heterogeneous, consisting of two distinct populations of precursors that are precommitted to become either CD1c+ or CD141+ cDCs. The two groups of lineage-primed precursors can be distinguished based on differential expression of CD172a. Both subpopulations of pre-cDCs arise in the adult bone marrow and can be found in cord blood and adult peripheral blood. Gene expression analysis revealed that CD172a+ and CD172a- pre-cDCs represent developmentally discrete populations that differentially express lineage-restricted transcription factors. A clinical trial of Flt3L injection revealed that this cytokine increases the number of both CD172a- and CD172a+ pre-cDCs in human peripheral blood.


Subject(s)
Antigens, CD1/metabolism , Antigens, Surface/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation/physiology , Glycoproteins/metabolism , Stem Cells/metabolism , Adult , Antigens, Differentiation/biosynthesis , Dendritic Cells/cytology , Humans , Receptors, Immunologic/biosynthesis , Stem Cells/cytology , Thrombomodulin , fms-Like Tyrosine Kinase 3/metabolism
17.
J Exp Med ; 213(13): 2931-2947, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27899441

ABSTRACT

The host responds to virus infection by activating type I interferon (IFN) signaling leading to expression of IFN-stimulated genes (ISGs). Dysregulation of the IFN response results in inflammatory diseases and chronic infections. In this study, we demonstrate that IFN regulatory factor 2 (IRF2), an ISG and a negative regulator of IFN signaling, influences alphavirus neuroinvasion and pathogenesis. A Sindbis virus strain that in wild-type (WT) mice only causes disease when injected into the brain leads to lethal encephalitis in Irf2-/- mice after peripheral inoculation. Irf2-/- mice fail to control virus replication and recruit immune infiltrates into the brain. Reduced B cells and virus-specific IgG are observed in the Irf2-/- mouse brains despite the presence of peripheral neutralizing antibodies, suggesting a defect in B cell trafficking to the central nervous system (CNS). B cell-deficient µMT mice are significantly more susceptible to viral infection, yet WT B cells and serum are unable to rescue the Irf2-/- mice. Collectively, our data demonstrate that proper localization of B cells and local production of antibodies in the CNS are required for protection. The work advances our understanding of host mechanisms that affect viral neuroinvasion and their contribution to immunity against CNS infections.


Subject(s)
Alphavirus Infections/immunology , B-Lymphocytes/immunology , Brain Diseases/immunology , Cell Movement/immunology , Interferon Regulatory Factor-2/immunology , Sindbis Virus/immunology , Alphavirus Infections/genetics , Alphavirus Infections/pathology , Animals , Antibodies, Viral/genetics , Antibodies, Viral/immunology , B-Lymphocytes/pathology , Brain Diseases/genetics , Brain Diseases/pathology , Brain Diseases/virology , Cell Movement/genetics , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interferon Regulatory Factor-2/genetics , Mice , Mice, Knockout
18.
Nat Protoc ; 10(9): 1407-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26292072

ABSTRACT

Human dendritic cells (DCs) develop from progressively restricted bone marrow (BM) progenitors: these progenitor cells include granulocyte, monocyte and DC progenitor (GMDP) cells; monocyte and DC progenitor (MDP) cells; and common DC progenitor (CDP) and DC precursor (pre-DC) cells. These four DC progenitors can be defined on the basis of the expression of surface markers such as CD34 and hematopoietin receptors. In this protocol, we describe five multiparametric flow cytometry panels that can be used as a tool (i) to simultaneously detect or phenotype the four DC progenitors, (ii) to isolate DC progenitors to enable in vitro differentiation or (iii) to assess the in vitro differentiation and proliferation of DC progenitors. The entire procedure from isolation of cells to flow cytometry can be completed in 3-7 h. This protocol provides optimized antibody panels, as well as gating strategies, for immunostaining of BM and cord blood specimens to study human DC hematopoiesis in health, disease and vaccine settings.


Subject(s)
Dendritic Cells/cytology , Flow Cytometry/methods , Stem Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Humans
19.
J Immunol Methods ; 425: 21-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26056939

ABSTRACT

Different dendritic cell (DC) subsets co-exist in humans and coordinate the immune response. Having a short life, DCs must be constantly replenished from their progenitors in the bone marrow through hematopoiesis. Identification of a DC-restricted progenitor in mouse has improved our understanding of how DC lineage diverges from myeloid and lymphoid lineages. However, identification of the DC-restricted progenitor in humans has not been possible because a system that simultaneously nurtures differentiation of human DCs, myeloid and lymphoid cells, is lacking. Here we report a cytokine and stromal cell culture that allows evaluation of CD34(+) progenitor potential to all three DC subsets as well as other myeloid and lymphoid cells, at a single cell level. Using this system, we show that human granulocyte-macrophage progenitors are heterogeneous and contain restricted progenitors to DCs.


Subject(s)
Dendritic Cells/immunology , Stem Cells/immunology , Stromal Cells/immunology , Antigens, CD34/immunology , Bone Marrow/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Cells, Cultured , Granulocyte-Macrophage Progenitor Cells/immunology , Hematopoiesis/immunology , Humans , Lymphocytes/immunology
20.
J Exp Med ; 212(3): 401-13, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25687281

ABSTRACT

Two subsets of conventional dendritic cells (cDCs) with distinct cell surface markers and functions exist in mouse and human. The two subsets of cDCs are specialized antigen-presenting cells that initiate T cell immunity and tolerance. In the mouse, a migratory cDC precursor (pre-CDC) originates from defined progenitors in the bone marrow (BM). Small numbers of short-lived pre-CDCs travel through the blood and replace cDCs in the peripheral organs, maintaining homeostasis of the highly dynamic cDC pool. However, the identity and distribution of the immediate precursor to human cDCs has not been defined. Using a tissue culture system that supports the development of human DCs, we identify a migratory precursor (hpre-CDC) that exists in human cord blood, BM, blood, and peripheral lymphoid organs. hpre-CDCs differ from premonocytes that are restricted to the BM. In contrast to earlier progenitors with greater developmental potential, the hpre-CDC is restricted to producing CD1c(+) and CD141(+) Clec9a(+) cDCs. Studies in human volunteers demonstrate that hpre-CDCs are a dynamic population that increases in response to levels of circulating Flt3L.


Subject(s)
Antigens, CD1/metabolism , Antigens, Surface/metabolism , Dendritic Cells/metabolism , Glycoproteins/metabolism , Cell Proliferation , Fetal Blood/cytology , Humans , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphoid Tissue/cytology , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Thrombomodulin , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL