Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Front Public Health ; 9: 616456, 2021.
Article in English | MEDLINE | ID: mdl-34195167

ABSTRACT

Introduction: Cervical cancer, a major consequence of persistent HPV infection, is the third most common cancer in women worldwide and has claimed around 311,000 women lives in 2018. The majority of these deaths took place in low- and middle-income countries (LMICs). In LMICs, where cervical cancer screening coverage is low, the HPV vaccine is a promising tool for preventing HPV infections and, thus, averting cervical cancer cases. In Indonesia, cervical cancer is the second most common cancer and HPV vaccination demonstration programs are underway in several provinces, but the HPV vaccine has not yet been introduced nationally. Since students are an important source of information for the community, and medical and nursing students are the future healthcare professionals, this study explored the knowledge, attitude, and acceptability of the HPV vaccine among University students in Indonesia. Methodology: A self-administered online questionnaire was used to assess the knowledge, attitude, and willingness of University students toward HPV vaccination. Result: A total of 433 students from Medical, Nursing, Social Sciences, and other faculties participated in the survey. It was identified that over 90% of the students were aware of cervical cancer and HPV, but only 68% knew about the HPV vaccine before participating in the study. Despite an average knowledge on the HPV vaccine, the students showed a strong willingness to receive the vaccine (95.8% acceptance rate). They believed that the HPV vaccine is safe and effective and that it will protect against HPV infection. The high cost and the lack of adequate information flow on HPV-related topics have been identified as potential barriers to the adoption of the HPV vaccine in Indonesia. Conclusion: Despite a high willingness for HPV vaccine uptake among students, there is a need to provide education on HPV vaccine-related topics to Indonesian students through awareness and training programs and improving the academic curriculum on vaccination for the long-term sustainability of the HPV vaccination program.


Subject(s)
Papillomavirus Vaccines , Uterine Cervical Neoplasms , Early Detection of Cancer , Female , Health Knowledge, Attitudes, Practice , Humans , Indonesia/epidemiology , Patient Acceptance of Health Care , Universities , Uterine Cervical Neoplasms/prevention & control , Vaccination
2.
Front Oncol ; 11: 780191, 2021.
Article in English | MEDLINE | ID: mdl-35127485

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasm characterised by the accumulation into granulomas of apoptosis-resistant pathological dendritic cells (LCH-DCs). LCH outcome ranges from self-resolving to fatal. Having previously shown that, (i) monocyte-derived DCs (Mo-DCs) from LCH patients differentiate into abnormal and pro-inflammatory IL-17A-producing DCs, and (ii) recombinant IL-17A induces survival and chemoresistance of healthy Mo-DCs, we investigated the link between IL-17A and resistance to apoptosis of LCH-DCs. In LCH granulomas, we uncovered the strong expression of BCL2A1 (alias BFL1), an anti-apoptotic BCL2 family member. In vitro, intracellular IL-17A expression was correlated with BCL2A1 expression and survival of Mo-DCs from LCH patients. Based on the chemotherapeutic drugs routinely used as first or second line LCH therapy, we treated these cells with vinblastine, or cytarabine and cladribine. Our preclinical results indicate that high doses of these drugs decreased the expression of Mcl-1, the main anti-apoptotic BCL2 family member for myeloid cells, and killed Mo-DCs from LCH patients ex vivo, without affecting BCL2A1 expression. Conversely, neutralizing anti-IL-17A antibodies decreased BCL2A1 expression, the downregulation of which lowered the survival rate of Mo-DCs from LCH patients. Interestingly, the in vitro combination of low-dose vinblastine with neutralizing anti-IL-17A antibodies killed Mo-DCs from LCH patients. In conclusion, we show that BCL2A1 expression induced by IL-17A links the inflammatory environment to the unusual pro-survival gene activation in LCH-DCs. Finally, these preclinical data support that targeting both Mcl-1 and BCL2A1 with low-dose vinblastine and anti-IL-17A biotherapy may represent a synergistic combination for managing recurrent or severe forms of LCH.

3.
MethodsX ; 7: 100997, 2020.
Article in English | MEDLINE | ID: mdl-32775222

ABSTRACT

Plasma IL-17A detection in Langerhans Cell Histiocytosis (LCH) is currently a source of debate. Indeed, 500-P07G (PeproTech) and 41802 (R&D Systems) anti-IL-17A antibodies have been suspected to recognize nonspecific proteins. To resolve this discrepancy, we set up two new ELISAs by using 41802 or neutralizing eBio64CAP17 (eBioscience) capture monoclonal antibodies that we compared to the commercial PeproTech ELISA kit. The three ELISAs, called E_500-P07G, E_41802 and E_eBio64CAP17, differ in their anti-IL-17A capture antibodies: either polyclonal, monoclonal or neutralizing monoclonal antibodies, respectively. Here, we show that these ELISAs had a similar capacity to specifically detect recombinant or native human IL-17A. However, a significantly lower plasma IL-17A detection was obtained with E_41802 compared to the two other ELISAs. Both E_500-P07G and E_eBio64CAP17 showed similar results. Consequently, we propose that the use of E_500-P07G and E_eBio64CAP17 may ensure more accurate and reliable results in the context of LCH studies. The highest plasma IL-17A levels in LCH patients compared to controls detected by both E_500-P07G and E_eBio64CAP17 ELISAs led us to propose these latter as reference techniques to investigate IL-17A as a potential new biomarker in LCH.•The customization of a new E_eBio64CAP17 ELISA is suitable to detect human IL-17A.•E_eBio64CAP17 ELISA protocol differs only in the anti-IL-17A capture antibody compared to the commercial E_500-P07G PeproTech kit.•Data generated using the E_eBio64CAP17 ELISA are consistent with the PeproTech kit.

4.
Cytokine ; 126: 154877, 2020 02.
Article in English | MEDLINE | ID: mdl-31629106

ABSTRACT

OBJECTIVE: Langerhans cell histiocytosis (LCH) is a granulomatous inflammatory myeloid neoplasia associated with a cytokine storm in both serum and lesions. Increased levels of plasma interleukin-17A (IL-17A) in LCH patients have been reported, but this finding was not confirmed in all studies. Neurodegeneration is a devastating complication of LCH (ND-LCH). We aimed to revisit the issue of plasma IL-17A levels in LCH, by using a larger number of patients, and also to investigate the relationship between IL-17A and LCH sequelae, especially ND-LCH. METHODS: Plasma samples from 68 LCH patients and 127 controls were analyzed for IL-17A levels by two ELISAs with different anti-IL-17A capture antibodies: either polyclonal or neutralizing monoclonal antibodies in 17polyAb-ELISA or 17mAb-ELISA, respectively. RESULTS: Both ELISAs had a similar capacity to specifically detect recombinant or native human IL-17A, as well as plasma IL-17A from LCH patients. We confirmed the finding of higher levels of plasma IL-17A in LCH patients compared to controls (p < 0.0001). The association of IL-17A with LCH was independent of the ELISA used, and of gender, age, disease class activity, and pattern of tissue-organ involvement (single-system versus multi-system). ROC analyses (p < 0.0001) allow to discriminate LCH patients from the control group, supporting the notion that IL-17A may be a potential biomarker for LCH. More interestingly, high IL-17A levels were significantly associated with LCH patients having sequelae, with the highest plasma levels in patients with ND-LCH (p < 0.0001). CONCLUSION: The association between high levels of IL-17A and LCH was confirmed. IL-17A may be associated with ND-LCH development. This might have therapeutic implications, offering a novel target for precision therapy of ND-LCH.


Subject(s)
Biomarkers/blood , Histiocytosis, Langerhans-Cell/blood , Interleukin-17/blood , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/complications , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Disease Progression , Female , Histiocytosis, Langerhans-Cell/complications , Humans , Infant , Inflammation , Male , Middle Aged
6.
J Lipid Res ; 56(6): 1110-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25833686

ABSTRACT

Interleukin 17A (IL-17A) is a proinflammatory cytokine involved in the pathogenesis of chronic inflammatory diseases. In the field of immunometabolism, we have studied the impact of IL-17A on the lipid metabolism of human in vitro-generated monocyte-derived dendritic cells (DCs). Microarrays and lipidomic analysis revealed an intense remodeling of lipid metabolism induced by IL-17A in DCs. IL-17A increased 2-12 times the amounts of phospholipids, cholesterol, triglycerides, and cholesteryl esters in DCs. Palmitic (16:0), stearic (18:0), and oleic (18:ln-9c) acid were the main fatty acid chains present in DCs. They were strongly increased in response to IL-17A while their relative proportion remained unchanged. Capture of extracellular lipids was the major mechanism of lipid droplet accumulation, visualized by electron microscopy and Oil Red O staining. Besides this foamy phenotype, IL-17A induced a mixed macrophage-DC phenotype and expression of the nuclear receptor NR1H3/liver X receptor-α, previously identified in the context of atherosclerosis as the master regulator of cholesterol homeostasis in macrophages. These IL-17A-treated DCs were as competent as untreated DCs to stimulate allogeneic naive T-cell proliferation. Following this first characterization of lipid-rich DCs, we propose to call these IL-17A-dependent cells "foamy DCs" and discuss the possible existence of foamy DCs in atherosclerosis, a metabolic and inflammatory disorder involving IL-17A.


Subject(s)
Atherosclerosis/metabolism , Dendritic Cells/metabolism , Fatty Acids/metabolism , Interleukin-17/metabolism , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Dendritic Cells/immunology , Foam Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Lipid Droplets/immunology , Lipid Droplets/metabolism , Lipid Metabolism/immunology , Liver X Receptors , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Blood ; 124(6): 867-72, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24894775

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare disease affecting people of any age, with widely variable clinical manifestations and different outcomes. The precise chain of events driving lesional granuloma formation has remained elusive for many years. There is evidence for inherited predisposition to and derangement of apoptosis and inflammation in lesional dendritic cells. Recently somatic BRAF(V600E) mutation in myeloid precursor dendritic cells was associated with the more aggressive form of the disease, although the same mutation in a more differentiated dendritic cell might drive a less aggressive disease. Whether this picture convincingly put LCH in the field of myeloid neoplasm remains to be determined. Altogether, these findings suggest that future therapeutic strategy might incorporate a screening of this genetic mutation for high-risk patients potentially suitable for target therapy.


Subject(s)
Histiocytosis, Langerhans-Cell , Amino Acid Substitution , Granuloma/pathology , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/therapy , Humans , Inflammation/pathology , Langerhans Cells/pathology , Models, Biological , Mutation, Missense , Prognosis , Proto-Oncogene Proteins B-raf/genetics
8.
Clin Immunol ; 153(1): 112-22, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24743018

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare disease of unknown cause with manifestations ranging from isolated granulomatous lesions to life-threatening multi-system organ involvement. This disorder is further characterized by infiltration of immune cells in affected tissues and an association with interleukin (IL)-17A has been reported. Here, we investigated the presence of IL-17A-producing cells among peripheral blood mononuclear cells isolated from LCH patients and observed a high percentage of IL-17A(+) monocytes in peripheral blood of LCH patients compared to controls. The IL-17A(+) monocytes were also positive for the transcription factor retinoic acid orphan receptor (ROR) γt and showed increased mRNA levels for both IL-17A and RORγt. Notably, IL-17A was produced by all monocyte subsets and the expression level was positively associated with LCH disease activity. These data support a role for monocytes in the pathogenesis of LCH. Future therapeutic approaches may consider identification of patients who may benefit from IL-17A-targeted interventions.


Subject(s)
Histiocytosis, Langerhans-Cell/immunology , Histiocytosis, Langerhans-Cell/metabolism , Interleukin-17/biosynthesis , Monocytes/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Gene Expression , Histiocytosis, Langerhans-Cell/genetics , Humans , Immunophenotyping , Infant , Interleukin-17/genetics , Leukocyte Count , Lipopolysaccharide Receptors/metabolism , Male , Monocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Transcription, Genetic , Young Adult
9.
Mol Biol Cell ; 25(3): 380-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284899

ABSTRACT

The function of osteoclasts (OCs), multinucleated giant cells (MGCs) of the monocytic lineage, is bone resorption. To resorb bone, OCs form podosomes. These are actin-rich adhesive structures that pattern into rings that drive OC migration and into "sealing-zones" (SZs) that confine the resorption lacuna. Although changes in actin dynamics during podosome patterning have been documented, the mechanisms that regulate these changes are largely unknown. From human monocytic precursors, we differentiated MGCs that express OC degradation enzymes but are unable to resorb the mineral matrix. We demonstrated that, despite exhibiting bona fide podosomes, these cells presented dysfunctional SZs. We then performed two-step differential transcriptomic profiling of bone-resorbing OCs versus nonresorbing MGCs to generate a list of genes implicated in bone resorption. From this list of candidate genes, we investigated the role of Rho/Rnd3. Using primary RhoE-deficient OCs, we demonstrated that RhoE is indispensable for OC migration and bone resorption by maintaining fast actin turnover in podosomes. We further showed that RhoE activates podosome component cofilin by inhibiting its Rock-mediated phosphorylation. We conclude that the RhoE-Rock-cofilin pathway, by promoting podosome dynamics and patterning, is central for OC migration, SZ formation, and, ultimately, bone resorption.


Subject(s)
Actins/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , rho GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Amides/pharmacology , Animals , Bone Resorption/genetics , Cattle , Cell Differentiation/genetics , Cell Movement , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Giant Cells/metabolism , Humans , Mice , Mice, Transgenic , Phosphorylation , Pyridines/pharmacology , Transcriptome , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
10.
Clin Dev Immunol ; 2013: 701305, 2013.
Article in English | MEDLINE | ID: mdl-23762095

ABSTRACT

Rheumatoid arthritis (RA) and Langerhans cell histiocytosis (LCH) are common and rare diseases, respectively. They associate myeloid cell recruitment and survival in inflammatory conditions with tissue destruction and bone resorption. Manipulating dendritic cell (DC), and, especially, regulating their half-life and fusion, is a challenge. Indeed, these myeloid cells display pathogenic roles in both diseases and may be an important source of precursors for differentiation of osteoclasts, the bone-resorbing multinucleated giant cells. We have recently documented that the proinflammatory cytokine IL-17A regulates long-term survival of DC by inducing BCL2A1 expression, in addition to the constitutive MCL1 expression. We summarize bibliography of the BCL2 family members and their therapeutic targeting, with a special emphasis on MCL1 and BCL2A1, discussing their potential impact on RA and LCH. Our recent knowledge in the survival pathway, which is activated to perform DC fusion in the presence of IL-17A, suggests that targeting MCL1 and BCL2A1 in infiltrating DC may affect the clinical outcomes in RA and LCH. The development of new therapies, interfering with MCL1 and BCL2A1 expression, to target long-term surviving inflammatory DC should be translated into preclinical studies with the aim to increase the well-being of patients with RA and LCH.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Dendritic Cells/drug effects , Histiocytosis, Langerhans-Cell/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/immunology , Myeloid Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Bone Resorption/immunology , Bone Resorption/pathology , Bone Resorption/prevention & control , Bone and Bones/drug effects , Bone and Bones/immunology , Bone and Bones/pathology , Cell Fusion , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation , Histiocytosis, Langerhans-Cell/immunology , Histiocytosis, Langerhans-Cell/pathology , Humans , Interleukin-17/pharmacology , Minor Histocompatibility Antigens , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cells/immunology , Myeloid Cells/pathology , Osteoclasts/drug effects , Osteoclasts/immunology , Osteoclasts/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction
11.
PLoS One ; 8(2): e56865, 2013.
Article in English | MEDLINE | ID: mdl-23441221

ABSTRACT

Dendritic cells initiate adaptive immune responses, leading either to control cancer by effector T cells or to exacerbate cancer by regulatory T cells that inhibit IFN-γ-mediated Th1-type response. Dendritic cells can also induce Th17-type immunity, mediated by IL-17A. However, the controversial role of this cytokine in cancer requires further investigations. We generated dendritic cells from peripheral blood monocytes to investigate lifespan, phenotype and chemoresistance of dendritic cells, treated with IL-17A with or without IFN-γ. Studying the expression of Bcl-2 family members, we demonstrated that dendritic cells constitutively express one pro-survival Bcl-2 member: MCL1. Immature dendritic cells were CD40(low)HLADR(low) CD1a(+) MCL1(+), did not express CD14, CD68 or BCL2A1, and displayed a short 2-day lifespan. IL-17A-treated DC exhibited a semi-mature (CD40(high) HLADR(low)) pre-M2 (CCL22(+) CD206(+) CD163(+) IL1RN(+) IL-10(-) CXCL10(-) IL-12(-)) mixed (CD1a(+) CD14+ CD68(+)) macrophage-dendritic cell phenotype. They efficiently exerted mannose receptor-mediated endocytosis and did not produce superoxide anions, in the absence of TLR engagement. Interestingly, IL-17A promoted a long-term survival of dendritic cells, beyond 12 days, that correlated to BCL2A1 induction, a pro-survival Bcl-2 family member. BCL2A1 transcription was activated by NF-κB, downstream of IL-17A transduction. Thus, immature dendritic cells only express MCL1, whereas IL-17A-treated dendritic cells concomitantly expressed two pro-survival Bcl-2 family members: MCL1 and BCL2A1. These latter developed chemoresistance to 11 of the 17 chemotherapy agents tested. However, high doses of either vinblastine or cytarabine decreased MCL1 expression and induced dendritic cell death. When IL-17A is produced in vivo, administration of anti-IL-17A biotherapy may impair dendritic cell survival by targeting BCL2A1 expression. Consequently, depending on the effector or regulatory role of dendritic cells, blocking IL-17A, may be either dangerous or beneficial for cancer outcomes, thus contributing to the apparent controversy around the role of IL-17A in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Drug Resistance, Neoplasm , Interleukin-17/pharmacology , Monocytes/cytology , Cell Differentiation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Interferon-gamma/pharmacology , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Phenotype , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic
12.
Nat Med ; 14(1): 81-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157139

ABSTRACT

IL-17A is a T cell-specific cytokine that is involved in chronic inflammations, such as Mycobacterium infection, Crohn's disease, rheumatoid arthritis and multiple sclerosis. Mouse models have explained the molecular basis of IL-17A production and have shown that IL-17A has a positive effect not only on granuloma formation and neurodegeneration through unknown mechanisms, but also on bone resorption through Receptor activator of NF-kappaB ligand (RANKL) induction in osteoblasts. Langerhans cell histiocytosis (LCH) is a rare disease of unknown etiology, lacking an animal model, that cumulates symptoms that are found separately in various IL-17A-related diseases, such as aggressive chronic granuloma formation, bone resorption and soft tissue lesions with occasional neurodegeneration. We examined IL-17A in the context of LCH and found that there were high serum levels of IL-17A during active LCH and unexpected IL-17A synthesis by dendritic cells (DCs), the major cell type in LCH lesions. We also found an IL-17A-dependent pathway for DC fusion, which was highly potentiated by IFN-gamma and led to giant cells expressing three major tissue-destructive enzymes: tartrate resistant acidic phosphatase and matrix metalloproteinases 9 and 12. IFN-gamma expression has been previously documented in LCH and observed in IL-17A-related diseases. Notably, serum IL-17A-dependent fusion activity correlates with LCH activity. Thus, IL-17A and IL-17A-stimulated DCs represent targets that may have clinical value in the treatment of LCH and other IL-17A-related inflammatory disorders.


Subject(s)
Dendritic Cells/metabolism , Histiocytosis, Langerhans-Cell/pathology , Interleukin-17/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Cell Fusion , Humans , Inflammation , Interferon-gamma/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Mice , Monocytes/metabolism , Mycobacterium/metabolism , Oligonucleotide Array Sequence Analysis
13.
Eur J Immunol ; 37(3): 747-57, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17304626

ABSTRACT

Dendritic cells (DC) are the mononuclear cells that initiate adaptive immune responses. Osteoclasts (OC) are the multinucleated giant cells that resorb bone. As previously described for human conventional DC (cDC), we demonstrate that murine cDC, either in vitro generated from Fms-like tyrosine kinase 3 (Flt3)+ bone marrow progenitors or ex vivo purified from spleen, are able to develop into OC in response to M-CSF and receptor activator of NF-kappaB ligand (RANKL) in vitro. This transdifferentiation is driven by the immune environment that controls cDC maturation, cell fusion, tartrate-resistant acid phosphatase (TRAP) and bone resorption activities. Only immature cDC have the capacity to become OC since mature cDC or plasmacytoid DC do not. Additions of the pro-inflammatory cytokines, such as IL-1beta and TNF-alpha, or human rheumatoid synovial fluid, increase murine cDC transdifferentiation into OC, whereas IFN-alpha inhibits it. The adaptive cytokine, IFN-gamma, inhibits cDC fusion while IL-4 increases it. IL-2, IFN-gamma and IL-4 inhibit TRAP and bone resorption activities contrary to IL-10, which enhances both activities. A putative new "immune multinucleated giant cell" unable to resorb bone, which is formed owing to IL-4, is underlined. The future analysis of cDC transdifferentiation into OC in murine models of inflammatory arthritis will give us the quantitative importance of this phenomenon in vivo.


Subject(s)
Cell Differentiation/immunology , Cytokines/physiology , Dendritic Cells/cytology , Growth Inhibitors/physiology , Osteoclasts/cytology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunity, Active , Immunity, Innate , Mice , Mice, Inbred C57BL , Osteoclasts/immunology , Osteoclasts/metabolism
14.
Mol Cell Proteomics ; 5(4): 726-36, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16428241

ABSTRACT

Dendritic cells (DCs) display the unique ability to activate naive T cells and to initiate primary T cell responses revealed in DC-T cell alloreactions. DCs frequently operate under stress conditions. Oxidative stress enhances the production of inflammatory cytokines by DCs. We performed a proteomic analysis to see which major changes occur, at the protein expression level, during DC differentiation and maturation. Comparative two-dimensional gel analysis of the monocyte, immature DC, and mature DC stages was performed. Manganese superoxide dismutase (Mn-SOD) reached 0.7% of the gel-displayed proteins at the mature DC stage. This important amount of Mn-SOD is a primary antioxidant defense system against superoxide radicals, but its product, H(2)O(2), is also deleterious for cells. Peroxiredoxin (Prx) enzymes play an important role in eliminating such peroxide. Prx1 expression level continuously increased during DC differentiation and maturation, whereas Prx6 continuously decreased, and Prx2 peaked at the immature DC stage. As a consequence, DCs were more resistant than monocytes to apoptosis induced by high amounts of oxidized low density lipoproteins containing toxic organic peroxides and hydrogen peroxide. Furthermore DC-stimulated T cells produced high levels of receptor activator of nuclear factor kappaB ligand, a chemotactic and survival factor for monocytes and DCs. This study provides insights into the original ability of DCs to express very high levels of antioxidant enzymes such as Mn-SOD and Prx1, to detoxify oxidized low density lipoproteins, and to induce high levels of receptor activator of nuclear factor kappaB ligand by the T cells they activate and further emphasizes the role that DCs might play in atherosclerosis, a pathology recognized as a chronic inflammatory disorder.


Subject(s)
Antioxidants/metabolism , Atherosclerosis/metabolism , Dendritic Cells/metabolism , Adult , Blotting, Western , Carrier Proteins/metabolism , Coculture Techniques , Dendritic Cells/enzymology , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Fluorescent Antibody Technique , Humans , Membrane Glycoproteins/metabolism , Peroxidases/metabolism , Peroxiredoxins , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Superoxide Dismutase/metabolism
15.
Blood ; 104(13): 4029-37, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15308576

ABSTRACT

Dendritic cells (DCs), the mononuclear cells that initiate immune response, and osteoclasts, the multinucleated bone-resorbing cells, are derived from monocyte/macrophage precursor cells. Granulocyte-macrophage colony-stimulating factor and macrophage colony-stimulating factor (M-CSF) reciprocally regulate the differentiation of both lineages in mice. Using human monocyte-derived DCs generated in vitro, we show that immature DCs transdifferentiate into functional osteoclasts (OCs) in the presence of M-CSF and receptor activator of nuclear factor-kappaB ligand (RANKL). Transdifferentiation operates through fusion of intermediate adherent bipolar fusiform mononuclear cells expressing CD14, CD1a, and RANKL and able to induce RANKL(+) T-cell proliferation. Surprisingly, DC fusion in vitro is faster and more efficient than monocyte fusion to form multinucleated giant cells. The transdifferentiation process reported here supports the existence of a high cellular plasticity within differentiated myeloid phagocytes. Importantly, this process is greatly enhanced by rheumatoid arthritis synovial fluid and involves proinflammatory cytokines such as interleukin 1 or tumor necrosis factor alpha, as well as components of the extracellular matrix such as hyaluronic acid. Our data therefore suggest that DC-derived OCs may be directly involved in the osteolytic lesions observed in human inflammatory bone diseases such as rheumatoid arthritis or in particular forms of Langerhans cell histiocytosis, characterized by accumulation of immature skin DCs and chronic lytic bone lesions.


Subject(s)
Arthritis, Rheumatoid/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Osteoclasts/cytology , T-Lymphocytes/immunology , Adult , Antigens, CD/blood , Arthritis, Rheumatoid/pathology , Cell Differentiation , Humans , Lymphocyte Activation , Osteoclasts/immunology , Reference Values
16.
BMC Immunol ; 3: 15, 2002 Oct 24.
Article in English | MEDLINE | ID: mdl-12398794

ABSTRACT

BACKGROUND: Macrophages, osteoclasts, dendritic cells, and microglia are highly specialized cells that belong to the mononuclear phagocyte system. Functional and phenotypic heterogeneity within the mononuclear phagocyte system may reveal differentiation plasticity of a common progenitor, but developmental pathways leading to such diversity are still unclear. RESULTS: Mouse bone marrow cells were expanded in vitro in the presence of Flt3-ligand (FL), yielding high numbers of non-adherent cells exhibiting immature monocyte characteristics. Cells expanded for 6 days, 8 days, or 11 days (day 6-FL, day 8-FL, and day 11-FL cells, respectively) exhibited constitutive potential towards macrophage differentiation. In contrast, they showed time-dependent potential towards osteoclast, dendritic, and microglia differentiation that was detected in day 6-, day 8-, and day 11-FL cells, in response to M-CSF and receptor activator of NFkappaB ligand (RANKL), granulocyte-macrophage colony stimulating-factor (GM-CSF) and tumor necrosis factor-alpha (TNFalpha), and glial cell-conditioned medium (GCCM), respectively. Analysis of cell proliferation using the vital dye CFSE revealed homogenous growth in FL-stimulated cultures of bone marrow cells, demonstrating that changes in differential potential did not result from sequential outgrowth of specific precursors. CONCLUSIONS: We propose that macrophages, osteoclasts, dendritic cells, and microglia may arise from expansion of common progenitors undergoing sequential differentiation commitment. This study also emphasizes differentiation plasticity within the mononuclear phagocyte system. Furthermore, selective massive cell production, as shown here, would greatly facilitate investigation of the clinical potential of dendritic cells and microglia.


Subject(s)
Dendritic Cells/physiology , Macrophages/physiology , Microglia/physiology , Osteoclasts/physiology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Stem Cells/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Membrane Glycoproteins/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Microglia/cytology , Osteoclasts/cytology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Stem Cells/cytology , Stem Cells/drug effects , Tumor Necrosis Factor-alpha/pharmacology , fms-Like Tyrosine Kinase 3
17.
J Virol ; 76(13): 6415-24, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12050353

ABSTRACT

In the thymus, epithelial cells comprise a heterogeneous population required for the generation of functional T lymphocytes, suggesting that thymic epithelium disruption by viruses may compromise T-cell lymphopoiesis in this organ. In a previous report, we demonstrated that in vitro, measles virus induced differentiation of cortical thymic epithelial cells as characterized by (i) cell growth arrest, (ii) morphological and phenotypic changes, and (iii) apoptotis as a final step of this process. In the present report, we have analyzed the mechanisms involved. First, measles virus-induced differentiation of thymic epithelial cells is shown to be strictly dependent on beta interferon (IFN-beta) secretion. In addition, transfection with double-stranded RNA, a common intermediate of replication for a broad spectrum of viruses, is reported to similarly mediate thymic epithelial cell differentiation through IFN-beta induction. Finally, we demonstrated that recombinant IFN-alpha, IFN-beta, or IFN-gamma was sufficient to induce differentiation and apoptosis of uninfected thymic epithelial cells. These observations suggested that interferon secretion by either infected cells or activated leukocytes, such as plasmacytoid dendritic cells or lymphocytes, may induce thymic epithelium disruption in a pathological context. Thus, we have identified a new mechanism that may contribute to thymic atrophy and altered T-cell lymphopoiesis associated with many infections.


Subject(s)
Epithelial Cells/cytology , Interferon-beta/physiology , Measles virus/physiology , Thymus Gland/cytology , Apoptosis , Cell Differentiation , Cell Line , Cells, Cultured , Epithelial Cells/virology , Humans , Interferon Type I/pharmacology , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/pharmacology , RNA, Double-Stranded/metabolism , Recombinant Proteins , Thymus Gland/virology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...