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










Publication year range
1.
Eur J Pediatr ; 183(3): 1425-1434, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38175262

ABSTRACT

Little information is available about the nature of the immune response in children after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. The aim of this study is to define the seroprevalence and the features of the antibody response in children of Southern Switzerland during the different waves of Coronavirus Disease 2019 (COVID-19) pandemic. By analyzing 756 sera collected from children aged 0 to 16 years admitted to the Institute of Pediatrics of Southern Switzerland during the prepandemic period (before March 2020) and the first four pandemic waves (between March 2020 and June 2022), we investigated binding titers, cross-reactivity, and neutralizing properties of the serum antibodies against SARS-CoV-2 variants. Seroprevalence varied from 6% during the first wave to 14% and 17% during the second and third waves, respectively, peaking at 39% during the fourth wave. The 96 seropositive cases were mostly asymptomatic (42.7%) or showed mild (20.8%) to moderate (32.3%) symptoms. Moderate symptoms and close contact with COVID-19-positive individuals were associated with a higher infection risk (P < 0.001). The antibody response was mainly driven by IgG directed to the receptor-binding domain (RBD) of Wuhan-1 SARS-CoV-2 Spike (S). Children infected in the first three waves produced antibodies with up to 11-fold and 5.5-fold reduction in binding and neutralizing titers, respectively, against different SARS-CoV-2 variants, including Beta, Delta, and Omicron BA.1, BA.2, and BA.5. Such reductions were less pronounced in children infected during the fourth wave, who showed the highest frequency and titers of neutralizing antibodies against the same variants. Compared to infection, vaccination with a Wuhan-1-based messenger RNA (mRNA) vaccine induced higher and heterogenous levels of antibodies cross-reacting to the different SARS-CoV-2 variants analyzed.   Conclusions: Despite the high burden of COVID-19 in Southern Switzerland, we observed an initial low seroprevalence of SARS-CoV-2 in children, which increased in the later waves. The antibody response was poor in the first three waves and improved in the fourth wave, when children produced higher levels of neutralizing antibodies after vaccination or infection with Delta and/or Omicron variants. What is Known: • Children were marginally affected by the initial SARS-CoV-2 variants. • The number of infected and hospitalized children increased after the appearance of the Omicron variants. What is New: • Seroprevalence of SARS-CoV-2 in children of Southern Switzerland increased overtime. • Children produced higher levels of neutralizing antibodies after vaccination or infection with Delta and/or Omicron variants in the fourth wave compared to children infected in the first three waves.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Child , COVID-19/epidemiology , COVID-19/prevention & control , Seroepidemiologic Studies , Switzerland/epidemiology , Antibodies, Neutralizing , Vaccination , Antibodies, Viral
2.
J Hepatol ; 79(5): 1129-1138, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37459920

ABSTRACT

BACKGROUND & AIMS: Chronic hepatitis B is a global public health problem, and coinfection with hepatitis delta virus (HDV) worsens disease outcome. Here, we describe a hepatitis B virus (HBV) surface antigen (HBsAg)-targeting monoclonal antibody (mAb) with the potential to treat chronic hepatitis B and chronic hepatitis D. METHODS: HBsAg-specific mAbs were isolated from memory B cells of HBV vaccinated individuals. In vitro neutralization was determined against HBV and HDV enveloped with HBsAg representing eight HBV genotypes. Human liver-chimeric mice were treated twice weekly with a candidate mAb starting 3 weeks post HBV inoculation (spreading phase) or during stable HBV or HBV/HDV coinfection (chronic phase). RESULTS: From a panel of human anti-HBs mAbs, VIR-3434 was selected and engineered for pre-clinical development. VIR-3434 targets a conserved, conformational epitope within the antigenic loop of HBsAg and neutralized HBV and HDV infection with higher potency than hepatitis B immunoglobulins in vitro. Neutralization was pan-genotypic against strains representative of HBV genotypes A-H. In the spreading phase of HBV infection in human liver-chimeric mice, a parental mAb of VIR-3434 (HBC34) prevented HBV dissemination and the increase in intrahepatic HBV RNA and covalently closed circular DNA. In the chronic phase of HBV infection or co-infection with HDV, HBC34 treatment decreased circulating HBsAg by >1 log and HDV RNA by >2 logs. CONCLUSIONS: The potently neutralizing anti-HBs mAb VIR-3434 reduces circulating HBsAg and HBV/HDV viremia in human liver-chimeric mice. VIR-3434 is currently in clinical development for treatment of patients with chronic hepatitis B or D. IMPACT AND IMPLICATIONS: Chronic infection with hepatitis B virus and co-infection with hepatitis D virus place approximately 290 million individuals worldwide at risk of severe liver disease and cancer. Available treatments result in low rates of functional cure or require lifelong therapy that does not eliminate the risk of liver disease. We isolated and characterized a potent human antibody that neutralizes hepatitis B and D viruses and reduces infection in a mouse model. This antibody could provide a new treatment for patients with chronic hepatitis B and D.

3.
J Infect Public Health ; 9(3): 231-5, 2016.
Article in English | MEDLINE | ID: mdl-27102927

ABSTRACT

The last century has witnessed the emergence of several previously unknown viruses as life-threatening human pathogens. Several examples include HIV, Ebola, Lujo, and, most recently, the Middle East respiratory syndrome (MERS) and Ebola. In this study, we describe a method for the swift generation of a human-derived monoclonal antibody, known as LCA60, as a treatment for MERS infections. LCA60 antibody was generated using the Cellclone Technology from the immortalized B cells of a human donor recovering from MERS. Only four months were required from the initial screening of B cells to the development of a stable CHO cell line suitable for the production of clinical grade antibody, thereby delineating a rapid pathway for the development of antiviral therapies against emerging viruses. Currently, the LCA60 antibody is being considered for clinical development, which includes prophylaxis in individuals at risk and a treatment for severe MERS-CoV infections.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Biotechnology/methods , Immunologic Factors/isolation & purification , Technology, Pharmaceutical/methods , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/pharmacology , CHO Cells , Coronavirus Infections/prevention & control , Coronavirus Infections/therapy , Cricetulus , Humans , Immunologic Factors/pharmacology , Immunotherapy/methods , Time Factors
4.
Eur J Dermatol ; 23(5): 618-28, 2013.
Article in English | MEDLINE | ID: mdl-24225049

ABSTRACT

BACKGROUND: The Toll-like receptor 7 (TLR7) activator imiquimod (IMQ) is safe and effective in treating actinic keratosis; however, an intermittent treatment regimen is necessary because of excessive local reactions. OBJECTIVES: To evaluate in vitro potency, pharmacodynamics/pharmacokinetics, toxicity and efficacy in vivo of the newly developed TLR7 ligand-phospholipid conjugate, TMX-202, in a gel formulation. MATERIAL AND METHODS: The effects of TMX-202 were assessed both in vitro on a murine macrophage cell line and in primary bone marrow-derived dendritic cells and in vivo on mice (C57BL/6-wild type, Myd88(-/-) and Tlr7(-/-)). RESULTS: TMX-202 was more potent than IMQ in vitro using murine and human cells. In contrast, in vivo it showed less systemic pro-inflammatory activity and better safety than IMQ. Moreover, the TMX-202 gel formulation exhibited at least comparable efficacy to Aldara in a mouse model for skin proliferative diseases. CONCLUSION: TMX-202 is safe and efficacious without causing excessive adverse effects, suggesting that it may be an alternative to Aldara for the treatment of proliferative skin conditions.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Glycerophospholipids/pharmacology , Glycerophospholipids/therapeutic use , Keratosis, Actinic/drug therapy , Membrane Glycoproteins/genetics , Toll-Like Receptor 7/genetics , Adenine/blood , Adenine/pharmacology , Adenine/therapeutic use , Aminoquinolines/blood , Aminoquinolines/pharmacology , Animals , Antineoplastic Agents/blood , Cell Line , Chemotactic Factors/blood , Dendritic Cells/physiology , Gels/pharmacology , Gels/therapeutic use , Glycerophospholipids/blood , Humans , Imiquimod , Interferon-gamma/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , Keratinocytes/physiology , Keratosis, Actinic/genetics , Leukocytes, Mononuclear/drug effects , Macrophages/physiology , Maximum Tolerated Dose , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Proto-Oncogene Proteins c-myc/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
5.
Gastroenterology ; 144(2): 346-356.e3, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23108068

ABSTRACT

BACKGROUND AND AIMS: Triggering receptor expressed on myeloid cells (TREM)-2 is a surface receptor detected on macrophages, dendritic cells, and microglia that binds repeated anionic motifs on yeast and Gram-positive and Gram-negative bacteria. Little is known about TREM-2 expression and function in the intestine or its role in inflammatory bowel disease (IBD). We investigated the expression of TREM-2 in the intestinal lamina propria and its role in the development of colonic inflammation. METHODS: We measured levels of TREM-2 in lamina propria mononuclear cells from surgical specimens collected from patients with IBD or cancer (controls). We analyzed the development of colitis in TREM-2 knockout and wild-type mice. Colon samples were isolated from mice and analyzed for cytokine expression, phagocytosis of bacteria, proliferation in colonic crypts, lamina propria mononuclear cell function, and T-cell activation by ovalbumin. RESULTS: TREM-2 was virtually absent from colon samples of control patients, but levels were significantly higher in within the inflamed mucosa of patients with IBD; it was mainly expressed by CD11c(+) cells. Levels of TREM-2 increased as acute or chronic colitis was induced in mice. TREM-2 knockout mice developed less severe colitis than wild-type mice; the knockout mice lost less body weight, had a lower disease activity index, and had smaller mucosal lesions in endoscopic analysis. Colon dendritic cells from TREM-2 knockout mice produced lower levels of inflammatory cytokines and had reduced levels of bacterial killing and T-cell activation than cells from wild-type mice. CONCLUSIONS: TREM-2 contributes to mucosal inflammation during development of colitis in mice. Levels of TREM-2 are increased within the inflamed mucosa of patients with IBD, indicating its potential as a therapeutic target.


Subject(s)
Colon/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/metabolism , Lymphocyte Activation/immunology , Membrane Glycoproteins/biosynthesis , Receptors, Immunologic/biosynthesis , Animals , Colon/immunology , Colon/pathology , Disease Models, Animal , Female , Flow Cytometry , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology
6.
J Neurochem ; 110(1): 284-94, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457124

ABSTRACT

Triggering receptor expressed in myeloid (TREM) cells 2, a receptor expressed by myeloid cells, osteoclasts and microglia, is known to play a protective role in bones and brain. Mutations of the receptor (or of its coupling protein, DAP12) sustain in fact a genetic disease affecting the two organs, the polycystic lipomembraneous osteodysplasia with sclerosing leukoencephalopathy (PLOSL or Nasu-Hakola disease). So far, specific agonist(s) of TREM2 have not been identified and its (their) transduction mechanisms are largely unknown. Heat shock protein 60 (Hsp60) is a mitochondrial chaperone that can also be harboured at the cell surface. By using constructs including the extracellular domain of TREM2 and the Fc domain of IgGs we have identified Hsp60 as the only TREM2-binding protein exposed at the surface of neuroblastoma N2A cells and astrocytes, and lacking in U373 astrocytoma. Treatment with Hsp60 was found to stimulate the best known TREM2-dependent process, phagocytosis, however, only in the microglial N9 cells rich in the receptor. Upon TREM2 down-regulation, the Hsp60-induced stimulation of N9 phagocytosis was greatly attenuated. Hsp60 is also released by many cell types, segregated within exosomes or shedding vesicles which might then undergo dissolution. However, the affinity of its binding (K(d) = 3.8 microM) might be too low for the soluble chaperone released from the vesicles to the extracellular space to induce a significant activation of TREM2. It might in contrast be appropriate for the binding of TREM2 to Hsp60 exposed at the surface of cells closely interacting with microglia. The ensuing stimulation of phagocytosis could play protective effects on the brain.


Subject(s)
Chaperonin 60/metabolism , Encephalitis/metabolism , Exocytosis/physiology , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Animals , Astrocytes/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Chaperonin 60/pharmacology , Down-Regulation/physiology , Encephalitis/physiopathology , Exocytosis/drug effects , Gliosis/metabolism , Gliosis/physiopathology , Humans , Immunoglobulin G/metabolism , Membrane Glycoproteins/agonists , Membrane Glycoproteins/chemistry , Mice , Microglia/drug effects , Molecular Chaperones/metabolism , Neurons/metabolism , Phagocytosis/drug effects , Phagocytosis/physiology , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats , Receptors, Cell Surface/agonists , Receptors, Cell Surface/chemistry , Receptors, Immunologic/agonists , Receptors, Immunologic/chemistry
7.
Brain ; 131(Pt 11): 3081-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18790823

ABSTRACT

Triggering receptor expressed on myeloid cells 2 (TREM-2) is a membrane-bound receptor expressed by microglia and macrophages. Engagement of TREM-2 on these cells has been reported to reduce inflammatory responses and, in microglial cells, to promote phagocytosis. TREM-2 function is critical within the CNS, as its genetic deficiency in humans causes neurodegeneration with myelin and axonal loss. Blockade of TREM-2 worsened the mouse model for multiple sclerosis. In the present study, a soluble form of TREM-2 protein has been identified by immunoprecipitation and by ELISA. Soluble TREM-2 protein (sTREM-2) was detected in human CSF, and was compared among subjects with relapsing-remitting multiple sclerosis (RR-MS; n = 52), primary progressive multiple sclerosis (PP-MS; n = 21), other inflammatory neurologic diseases (OIND; n = 19), and non-inflammatory neurologic diseases (NIND; n = 41). Compared to NIND subjects, CSF sTREM-2 levels were significantly higher in RR-MS (P = 0.004 by ANOVA) and PP-MS (P < 0.001) subjects, as well as in OIND (P < 0.001) subjects. In contrast, levels of sTREM-2 in blood did not differ among the groups. Furthermore, TREM-2 was detected on a subset of CSF monocytes by flow cytometry, and was also highly expressed on myelin-laden macrophages in eight active demyelinating lesions from four autopsied multiple sclerosis subjects. The elevated levels of sTREM-2 in CSF of multiple sclerosis patients may inhibit the anti-inflammatory function of the membrane-bound receptor suggesting sTREM-2 to be a possible target for future therapies.


Subject(s)
Encephalomyelitis/cerebrospinal fluid , Membrane Glycoproteins/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Adolescent , Adult , Cells, Cultured , Dendritic Cells/metabolism , Encephalomyelitis/blood , Encephalomyelitis/pathology , Enzyme-Linked Immunosorbent Assay/methods , Female , Foam Cells/metabolism , Foam Cells/pathology , Humans , Male , Membrane Glycoproteins/blood , Middle Aged , Monocytes/metabolism , Multiple Sclerosis/blood , Multiple Sclerosis/pathology , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Multiple Sclerosis, Chronic Progressive/pathology , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/pathology , Pons/metabolism , Pons/pathology , Receptors, Immunologic/blood , Young Adult
8.
EMBO J ; 26(5): 1292-302, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17304212

ABSTRACT

Interleukin-12 (IL-12) is a key cytokine for the development of T helper type 1 (Th1) responses; however, naïve CD4(+) T cells do not express IL-12Rbeta2, and are therefore unresponsive to IL-12. We have examined the mechanisms that control Th1-specific expression of the human IL-12Rbeta2 gene at early time points after T-cell stimulation. We have identified a Th1-specific enhancer element that binds signal transducer and activator of transcription 4 (STAT4) in vivo in developing Th1 but not Th2 cells. T-cell receptor (TCR) signaling induced histone hyperacetylation and recruitment of BRG1, the ATPase subunit of the SWI/SNF-like BAF chromatin remodeling complex, to the IL-12Rbeta2 regulatory regions and was associated with low-level gene transcription at the IL-12Rbeta2 locus. However, high-level IL-12Rbeta2 expression required TCR triggering in the presence of IL-12. Our results indicate a synergistic role of TCR-induced chromatin remodeling and cytokine-induced STAT4 activation to direct IL-12Rbeta2 expression during Th1 cell development.


Subject(s)
Cell Differentiation/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Receptors, Interleukin-12/genetics , STAT4 Transcription Factor/genetics , Th1 Cells/metabolism , Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Humans , Interferon-alpha/pharmacology , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Interleukin-12/pharmacology , Interleukin-4/pharmacology , Models, Biological , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Interference , Receptors, Interleukin-12/immunology , Receptors, Interleukin-12/metabolism , STAT4 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Th1 Cells/cytology , Th1 Cells/immunology , Transcription Factors/genetics
9.
J Steroid Biochem Mol Biol ; 103(3-5): 277-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17254779

ABSTRACT

The searches for drugs that exhibit antineoplastic activity and regulate blood pressure are among the most prevalent and compelling research activities today. Amazingly, there is ample precedence for the antiproliferative action of vitamin-D-related compounds and their role as endocrine suppressors of renin biosynthesis. We have recently synthesized a number of novel calcitriol analogs of the gemini family and originally selected for further studies an epimeric pair related to 19-nor-calcitriol whose 21-methyl group was replaced by a 5,5,5-trifluoro-4-hydroxy-4-(trifluoromethyl)-2-pentynyl group. While maintaining the acceptable calcemic responses, the IC50 concentrations of interferon-gamma release were reduced and the antiproliferative activity and inhibition of renin mRNA expression enhanced. Replacing the geminal methyl groups on the calcitriol-related side chain of these gemini compounds with trideuteriomethyl moieties further boosted the potency in the colon cancer model in mice some 10-fold, reduced NMU-induced breast cancer carcinogenesis in rats and decreased the IC(50) values for renin mRNA inhibition into the pM range.


Subject(s)
Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Neoplasms/genetics , Neoplasms/pathology , Renin/genetics , Animals , Body Weight/drug effects , Calcitriol/chemical synthesis , Calcitriol/chemistry , Calcium/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Hydroxylation , Interferon-gamma/metabolism , Mice , Molecular Structure , Neoplasm Transplantation , Neoplasms/metabolism , RNA, Messenger/genetics , Rats , Renin/metabolism
10.
Infect Immun ; 74(5): 2823-30, 2006 May.
Article in English | MEDLINE | ID: mdl-16622220

ABSTRACT

The triggering receptor expressed on myeloid cell type 1 (TREM-1) is a cell surface molecule that has been identified on both human and murine polymorphonuclear neutrophils and mature monocytes. The activation of TREM-1 in the presence of microbial components amplifies the inflammatory response and may be responsible for the hyperresponsiveness observed during the initial stage of sepsis. To investigate the effect of the modulation of the TREM-1 pathway during experimental murine sepsis, we used analogue synthetic peptides derived from the extracellular moiety of TREM-1. The TREM-1 ligand was expressed on both peritoneal and peripheral neutrophils during experimental peritonitis in mice. The TREM-1 peptides inhibited the recognition by TREM-1 of its ligand and protected endotoxinic mice from death. In septic rats, the TREM-1 peptides improved the hemodynamic status, attenuated the development of lactic acidosis, modulated the production of such proinflammatory cytokines as tumor necrosis factor alpha and interleukin-1beta, and improved survival. The protective effect of these peptides on arterial pressure could partly be explained by a decreased production of nitric oxide. These data suggest that in vivo modulation of TREM-1 might be a suitable therapeutic tool for the treatment of sepsis.


Subject(s)
Membrane Glycoproteins/physiology , Receptors, Immunologic/physiology , Shock, Septic/immunology , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , Disease Models, Animal , Hydrogen-Ion Concentration , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nitric Oxide/physiology , Rats , Rats, Wistar , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...