Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 1379, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35296643

ABSTRACT

Anti tumour necrosis factor (anti-TNF) drugs increase the risk of serious respiratory infection and impair protective immunity following pneumococcal and influenza vaccination. Here we report SARS-CoV-2 vaccine-induced immune responses and breakthrough infections in patients with inflammatory bowel disease, who are treated either with the anti-TNF antibody, infliximab, or with vedolizumab targeting a gut-specific anti-integrin that does not impair systemic immunity. Geometric mean [SD] anti-S RBD antibody concentrations are lower and half-lives shorter in patients treated with infliximab than vedolizumab, following two doses of BNT162b2 (566.7 U/mL [6.2] vs 4555.3 U/mL [5.4], p <0.0001; 26.8 days [95% CI 26.2 - 27.5] vs 47.6 days [45.5 - 49.8], p <0.0001); similar results are also observed with ChAdOx1 nCoV-19 vaccination (184.7 U/mL [5.0] vs 784.0 U/mL [3.5], p <0.0001; 35.9 days [34.9 - 36.8] vs 58.0 days [55.0 - 61.3], p value < 0.0001). One fifth of patients fail to mount a T cell response in both treatment groups. Breakthrough SARS-CoV-2 infections are more frequent (5.8% (201/3441) vs 3.9% (66/1682), p = 0.0039) in patients treated with infliximab than vedolizumab, and the risk of breakthrough SARS-CoV-2 infection is predicted by peak anti-S RBD antibody concentration after two vaccine doses. Irrespective of the treatments, higher, more sustained antibody levels are observed in patients with a history of SARS-CoV-2 infection prior to vaccination. Our results thus suggest that adapted vaccination schedules may be required to induce immunity in at-risk, anti-TNF-treated patients.


Subject(s)
COVID-19 , Inflammatory Bowel Diseases , Viral Vaccines , Antibodies, Monoclonal, Humanized/therapeutic use , BNT162 Vaccine , COVID-19 Vaccines , ChAdOx1 nCoV-19 , Humans , Inflammatory Bowel Diseases/drug therapy , Infliximab/therapeutic use , SARS-CoV-2 , T-Lymphocytes , Tumor Necrosis Factor Inhibitors
2.
J Infect Dis ; 215(9): 1480-1487, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28329119

ABSTRACT

Background: Recently acquired and remotely acquired latent Mycobacterium tuberculosis infection (LTBI) are clinically indistinguishable, yet recent acquisition of infection is the greatest risk factor for progression to tuberculosis in immunocompetent individuals. We aimed to evaluate the ability of cellular immune signatures that differ between active tuberculosis and LTBI to distinguish recently from remotely acquired LTBI. Methods: Fifty-nine individuals were recruited: 20 had active tuberculosis, 19 had recently acquired LTBI, and 20 had remotely acquired LTBI. The proportion of mycobacteria-specific CD4+ T cells secreting tumor necrosis factor α (TNF-α) but not interferon γ or interleukin 2 which had a differentiated effector phenotype (TNF-α-only TEFF), and the level of CD27 expression on IFN-γ-producing CD4+ T cells, were detected by flow cytometry. Results: The TNF-α-only TEFF signature was significantly higher in the group with recently acquired LTBI, compared with the group with remotely acquired LTBI (P < .0001), and it discriminated between these groups with high sensitivity and specificity, with an area under the curve of 0.87. Two signatures incorporating CD27 expression did not distinguish between recently and remotely acquired LTBI. Interestingly, the TNF-α-only TEFF signature in participants with recently acquired LTBI was more similar to that in participants with tuberculosis than that in participants with remotely acquired LTBI, suggesting that recently acquired LTBI is immunologically more similar to tuberculosis than remotely acquired LTBI. Conclusions: These findings reveal marked biological heterogeneity underlying the clinically homogeneous phenotype of LTBI, providing a rationale for immunological risk stratification to improve targeting of LTBI treatment.


Subject(s)
Latent Tuberculosis/epidemiology , Latent Tuberculosis/immunology , Mycobacterium tuberculosis/immunology , Adult , Aged , Biomarkers/blood , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Interferon-gamma/blood , Interleukin-2/blood , Latent Tuberculosis/diagnosis , Male , Middle Aged , Prospective Studies , Sensitivity and Specificity , Tumor Necrosis Factor-alpha/blood , Young Adult
3.
Invest Ophthalmol Vis Sci ; 53(2): 888-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22247464

ABSTRACT

PURPOSE: Noninfectious uveitis is characterized by a dysregulated inflammatory or immune response in the eye. It is unclear whether this represents a failure of immune privilege or an overwhelming inflammatory drive that has exceeded the capacity of regulatory mechanisms that are still functioning. The authors investigated immune regulation in the human eye during intraocular inflammation (uveitis) and its impact on dendritic cell (DC) function and subsequent T-cell responses. METHODS: Myeloid DCs were isolated from the aqueous humor (AqH) and peripheral blood of patients with active uveitis and characterized by flow cytometry. The effect of uveitis AqH was interrogated in an in vitro model of peripheral blood monocyte-derived DCs from healthy controls. RESULTS: Myeloid DCs isolated from uveitic AqH were characterized by elevated major histocompatibility complex classes I and II (MHC I/II), but reduced CD86 compared with matched peripheral blood DCs. Exposure of peripheral blood monocyte-derived DCs from healthy controls to the inflammatory AqH supernatant recapitulated this phenotype. Despite interferon gamma (IFNγ)-dependent upregulation of MHC I, inflammatory AqH was overall suppressive to DC function, with reduced CD86 expression and diminished T-cell responses. This suppressive effect was equal to or greater than that induced by noninflammatory AqH, but was glucocorticoid independent (in contrast to noninflammatory AqH). CONCLUSIONS: These data indicate that the ocular microenvironment continues to regulate DC function during uveitis, despite IFNγ-driven upregulation of MHC expression, supporting the hypothesis that immune regulation within the eye is maintained during inflammation.


Subject(s)
Aqueous Humor/physiology , Dendritic Cells/physiology , Immunity, Cellular , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Uveitis/immunology , Aqueous Humor/cytology , Cells, Cultured , Dendritic Cells/immunology , Flow Cytometry , Humans , Uveitis/metabolism , Uveitis/pathology
4.
J Immunol ; 186(1): 305-11, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106846

ABSTRACT

Aqueous humor (AqH) has been shown to have significant immunosuppressive effects on APCs in animal models. We wanted to establish whether, in humans, AqH can regulate dendritic cell (DC) function and to identify the dominant mechanism involved. Human AqH inhibited the capacity of human peripheral blood monocyte-derived DC to induce naive CD4(+) T cell proliferation and cytokine production in vitro, associated with a reduction in DC expression of the costimulatory molecule CD86. This was seen both for DC cultured under noninflammatory conditions (immature DC) and for DC stimulated by proinflammatory cytokines (mature DC). DC expression of MHC classes I/II and CD83 was reduced (mature DC only). Myeloid DC from peripheral blood were similarly sensitive to the effects of human AqH, but only under inflammatory conditions. The addition of α-melanocyte stimulating hormone and vasoactive intestinal peptide did not cause significant inhibition at physiological levels. However, the addition of exogenous cortisol at physiological levels recapitulated the AqH-induced reduction in CD86 and inhibition of DC-induced T cell proliferation, and blockade of cortisol in AqH partially reversed its suppressive effects. TGF-ß2 had an additional effect with cortisol, and although simultaneous blockade of cortisol and TGF-ß2 in AqH reduced its effectiveness, there was still a cortisol- and TGF-ß-independent component. In humans, AqH regulates DC maturation and function by the combined actions of cortisol and TGF-ß2, a pathway that is likely to contribute to the maintenance of immune privilege in the eye.


Subject(s)
Aqueous Humor/immunology , Dendritic Cells/immunology , Eye/immunology , Hydrocortisone/physiology , Immune Tolerance , Transforming Growth Factor beta2/physiology , Antigen Presentation/immunology , Aqueous Humor/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/metabolism , Eye/metabolism , HLA Antigens/biosynthesis , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class II/biosynthesis , Humans , Hydrocortisone/antagonists & inhibitors , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transforming Growth Factor beta2/antagonists & inhibitors
5.
Genes Chromosomes Cancer ; 48(11): 983-92, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19681120

ABSTRACT

Cervical cancer (CC) is the second most common cancer in women. Currently, no tractable molecular-based therapeutic targets exist for patients with invasive CC and no predictive markers of risk assessment for progression of precancerous lesions are identified. New molecular insights into CC pathogenesis are urgently needed. Towards this goal, we first determined the copy number alterations of chromosome 4 and then examined the role of PCDH10 mapped to 4q28 as a candidate tumor suppressor gene. We identified monosomy 4 in 47% of 81 invasive CC studied by SNP array and found that 91% of 130 invasive CC harboring methylation in the promoter region of the PCDH10 gene. We then showed that aberrant promoter hypermethylation of PCDH10 is associated with downregulation of gene expression and cell lines exposed to demethylating agent resulted in profound reactivated gene expression. We also showed that the promoter methylation in the PCDH10 gene occurs at an earliest identifiable stage of low-grade squamous intraepithelial lesion. Our studies demonstrate that inactivation of PCDH10 may be a critical event in CC progression and form a potentially useful therapeutic target for CC.


Subject(s)
Cadherins/genetics , DNA Methylation , Uterine Cervical Neoplasms/genetics , Cadherins/metabolism , Cell Line, Tumor , Chromosomes, Human, Pair 4 , DNA Copy Number Variations , DNA Mutational Analysis , Disease Progression , Epigenesis, Genetic , Female , Gene Dosage , Gene Expression , Humans , Monosomy , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protocadherins , Uterine Cervical Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...