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










Publication year range
1.
Front Immunol ; 15: 1369436, 2024.
Article in English | MEDLINE | ID: mdl-38629062

ABSTRACT

Adenovirus vaccines, particularly the COVID-19 Ad5-nCoV adenovirus vaccine, have emerged as promising tools in the fight against infectious diseases. In this study, we investigated the structure of the T cell response to the Spike protein of the SARS-CoV-2 virus used in the COVID-19 Ad5-nCoV adenoviral vaccine in a phase 3 clinical trial (NCT04540419). In 69 participants, we collected peripheral blood samples at four time points after vaccination or placebo injection. Sequencing of T cell receptor repertoires from Spike-stimulated T cell cultures at day 14 from 17 vaccinated revealed a more diverse CD4+ T cell repertoire compared to CD8+. Nevertheless, CD8+ clonotypes accounted for more than half of the Spike-specific repertoire. Our longitudinal analysis showed a peak T cell response at day 14, followed by a decline until month 6. Remarkably, multiple T cell clonotypes persisted for at least 6 months after vaccination, as demonstrated by ex vivo stimulation. Examination of CDR3 regions revealed homologous sequences in both CD4+ and CD8+ clonotypes, with major CD8+ clonotypes sharing high similarity with annotated sequences specific for the NYNYLYRLF peptide, suggesting potential immunodominance. In conclusion, our study demonstrates the immunogenicity of the Ad5-nCoV adenoviral vaccine and highlights its ability to induce robust and durable T cell responses. These findings provide valuable insight into the efficacy of the vaccine against COVID-19 and provide critical information for ongoing efforts to control infectious diseases.


Subject(s)
COVID-19 , Communicable Diseases , Vaccines , Humans , COVID-19 Vaccines , Spike Glycoprotein, Coronavirus , COVID-19/prevention & control , SARS-CoV-2 , T-Lymphocytes , Adenoviridae/genetics
2.
Cancer Discov ; 12(8): 1960-1983, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35723626

ABSTRACT

Although inflammatory mechanisms driving hepatocellular carcinoma (HCC) have been proposed, the regulators of anticancer immunity in HCC remain poorly understood. We found that IL27 receptor (IL27R) signaling promotes HCC development in vivo. High IL27EBI3 cytokine or IL27RA expression correlated with poor prognosis for patients with HCC. Loss of IL27R suppressed HCC in vivo in two different models of hepatocarcinogenesis. Mechanistically, IL27R sig-naling within the tumor microenvironment restrains the cytotoxicity of innate cytotoxic lymphocytes. IL27R ablation enhanced their accumulation and activation, whereas depletion or functional impairment of innate cytotoxic cells abrogated the effect of IL27R disruption. Pharmacologic neutralization of IL27 signaling increased infiltration of innate cytotoxic lymphocytes with upregulated cytotoxic molecules and reduced HCC development. Our data reveal an unexpected role of IL27R signaling as an immunologic checkpoint regulating innate cytotoxic lymphocytes and promoting HCC of different etiologies, thus indicating a therapeutic potential for IL27 pathway blockade in HCC. SIGNIFICANCE: HCC, the most common form of liver cancer, is characterized by a poor survival rate and limited treatment options. The discovery of a novel IL27-dependent mechanism controlling anticancer cytotoxic immune response will pave the road for new treatment options for this devastating disease. This article is highlighted in the In This Issue feature, p. 1825.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Interleukin-27 , Liver Neoplasms , T-Lymphocytes, Cytotoxic , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Interleukin-27/immunology , Interleukins/immunology , Liver Neoplasms/immunology , Prognosis , Receptors, Interleukin/immunology , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology
3.
Clin Infect Dis ; 75(1): e1-e9, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35435222

ABSTRACT

BACKGROUND: During the ongoing coronavirus disease 2019 (COVID-19) pandemic, many individuals were infected with and have cleared the virus, developing virus-specific antibodies and effector/memory T cells. An important unanswered question is what levels of T-cell and antibody responses are sufficient to protect from the infection. METHODS: In 5340 Moscow residents, we evaluated anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin M (IgM)/immunoglobulin G (IgG) titers and frequencies of the T cells specific to the membrane, nucleocapsid, and spike proteins of SARS-CoV-2, using interferon gamma (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assay. Additionally, we evaluated the fractions of virus-specific CD4+ and CD8+ T cells using intracellular staining of IFN-γ and interleukin 2 followed by flow cytometry. We analyzed the COVID-19 rates as a function of the assessed antibody and T-cell responses, using the Kaplan-Meier estimator method, for up to 300 days postinclusion. RESULTS: We showed that T-cell and antibody responses are closely interconnected and are commonly induced concurrently. Magnitudes of both responses inversely correlated with infection probability. Individuals positive for both responses demonstrated the highest levels of protectivity against the SARS-CoV-2 infection. A comparable level of protection was found in individuals with antibody response only, whereas the T-cell response by itself granted only intermediate protection. CONCLUSIONS: We found that the contribution of the virus-specific antibodies to protection against SARS-CoV-2 infection is more pronounced than that of the T cells. The data on the virus-specific IgG titers may be instructive for making decisions in personalized healthcare and public anti-COVID-19 policies. Clinical Trials Registration. NCT04898140.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Humans , Immunoglobulin G , Prospective Studies
4.
J Immunol ; 208(5): 1139-1145, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35101893

ABSTRACT

Despite measures taken world-wide, the coronavirus disease 2019 (COVID-19) pandemic continues. Because efficient antiviral drugs are not yet widely available, vaccination is the best option to control the infection rate. Although this option is obvious in the case of COVID-19-naive individuals, it is still unclear when individuals who have recovered from a previous SARS-CoV-2 infection should be vaccinated and whether the vaccination raises immune responses against the coronavirus and its novel variants. In this study, we collected peripheral blood from 84 healthy human donors of different COVID-19 status who were vaccinated with the Sputnik Light vaccine and measured the dynamics of the Ab and T cell responses, as well as the virus-neutralizing activity (VNA) in serum, against two SARS-CoV-2 variants, B.1.1.1 and B.1.617.2. We showed that vaccination of individuals previously exposed to the virus considerably boosts the existing immune response. In these individuals, receptor-binding domain (RBD)-specific IgG titers and VNA in serum were already elevated on the 7th day after vaccination, whereas COVID-19-naive individuals developed the Ab response and VNA mainly 21 d postvaccination. Additionally, we found a strong correlation between RBD-specific IgG titers and VNA in serum, and according to these data vaccination may be recommended when the RBD-specific IgG titers drop to 142.7 binding Ab units/ml or below. In summary, the results of the study demonstrate that vaccination is beneficial for both COVID-19-naive and recovered individuals, especially since it raises serum VNA against the B.1.617.2 variant, one of the five SARS-CoV-2 variants of concern.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Protein Domains/immunology , Russia , T-Lymphocytes/immunology , Vaccination
5.
Int J Mol Sci ; 24(1)2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36613860

ABSTRACT

The clinical course of the new coronavirus disease 2019 (COVID-19) has shown that patients with chronic lymphocytic leukemia (CLL) are characterized by a high mortality rate, poor response to standard treatment, and low virus-specific antibody response after recovery and/or vaccination. To date, there are no data on the safety and efficacy of the combined vector vaccine Sputnik V in patients with CLL. Here, we analyzed and compared the magnitudes of the antibody and T cell responses after vaccination with the Sputnik V vaccine among healthy donors and individuals with CLL with different statuses of preexposure to coronavirus. We found that vaccination of the COVID-19-recovered individuals resulted in the boosting of pre-existing immune responses in both healthy donors and CLL patients. However, the COVID-19-naïve CLL patients demonstrated a considerably lower antibody response than the healthy donors, although they developed a robust T cell response. Regardless of the previous infection, the individuals over 70 years old demonstrated a decreased response to vaccination, as did those receiving anti-CD20 therapy. In summary, we showed that Sputnik V, like other vaccines, did not induce a robust antibody response in individuals with CLL; however, it provided for the development of a significant anti-COVID-19 T cell response.


Subject(s)
COVID-19 Vaccines , COVID-19 , Leukemia, Lymphocytic, Chronic, B-Cell , Aged , Humans , Antibodies, Viral , COVID-19/prevention & control , T-Lymphocytes , Vaccination , Vaccines, Combined , COVID-19 Vaccines/immunology , Vaccines, Synthetic
6.
Immunity ; 53(6): 1245-1257.e5, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326767

ABSTRACT

Understanding the hallmarks of the immune response to SARS-CoV-2 is critical for fighting the COVID-19 pandemic. We assessed antibody and T cell reactivity in convalescent COVID-19 patients and healthy donors sampled both prior to and during the pandemic. Healthy donors examined during the pandemic exhibited increased numbers of SARS-CoV-2-specific T cells, but no humoral response. Their probable exposure to the virus resulted in either asymptomatic infection without antibody secretion or activation of preexisting immunity. In convalescent patients, we observed a public and diverse T cell response to SARS-CoV-2 epitopes, revealing T cell receptor (TCR) motifs with germline-encoded features. Bulk CD4+ and CD8+ T cell responses to the spike protein were mediated by groups of homologous TCRs, some of them shared across multiple donors. Overall, our results demonstrate that the T cell response to SARS-CoV-2, including the identified set of TCRs, can serve as a useful biomarker for surveying antiviral immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Epitopes, T-Lymphocyte/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/metabolism , Adolescent , Adult , Antibodies, Viral/metabolism , Asymptomatic Infections , Cells, Cultured , Convalescence , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunity , Immunologic Memory , Lymphocyte Activation , Male , Middle Aged , Pandemics , Receptors, Antigen, T-Cell/metabolism , Spike Glycoprotein, Coronavirus/immunology , Young Adult
7.
Nat Commun ; 10(1): 5046, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31695038

ABSTRACT

Abdominal aortic aneurysm (AAA) is a prevalent life-threatening disease, where aortic wall degradation is mediated by accumulated immune cells. Although cytokines regulate inflammation within the aorta, their contribution to AAA via distant alterations, particularly in the control of hematopoietic stem cell (HSC) differentiation, remains poorly defined. Here we report a pathogenic role for the interleukin-27 receptor (IL-27R) in AAA, as genetic ablation of IL-27R protects mice from the disease development. Mitigation of AAA is associated with a blunted accumulation of myeloid cells in the aorta due to the attenuation of Angiotensin II (Ang II)-induced HSC expansion. IL-27R signaling is required to induce transcriptional programming to overcome HSC quiescence and increase differentiation and output of mature myeloid cells in response to stress stimuli to promote their accumulation in the diseased aorta. Overall, our studies illuminate how a prominent vascular disease can be distantly driven by a cytokine-dependent regulation of bone marrow precursors.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Interleukin-27/metabolism , Myelopoiesis/physiology , Receptors, Interleukin/metabolism , Aneurysm/metabolism , Angiotensin II/metabolism , Animals , Aorta/pathology , Aortic Aneurysm, Abdominal/pathology , Blood Pressure , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Myeloid Cells/pathology , Receptors, Interleukin/genetics , Signal Transduction
8.
Immunity ; 49(5): 943-957.e9, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30389414

ABSTRACT

Although commensal flora is involved in the regulation of immunity, the interplay between cytokine signaling and microbiota in atherosclerosis remains unknown. We found that interleukin (IL)-23 and its downstream target IL-22 restricted atherosclerosis by repressing pro-atherogenic microbiota. Inactivation of IL-23-IL-22 signaling led to deterioration of the intestinal barrier, dysbiosis, and expansion of pathogenic bacteria with distinct biosynthetic and metabolic properties, causing systemic increase in pro-atherogenic metabolites such as lipopolysaccharide (LPS) and trimethylamine N-oxide (TMAO). Augmented disease in the absence of the IL-23-IL-22 pathway was mediated in part by pro-atherogenic osteopontin, controlled by microbial metabolites. Microbiota transfer from IL-23-deficient mice accelerated atherosclerosis, whereas microbial depletion or IL-22 supplementation reduced inflammation and ameliorated disease. Our work uncovers the IL-23-IL-22 signaling as a regulator of atherosclerosis that restrains expansion of pro-atherogenic microbiota and argues for informed use of cytokine blockers to avoid cardiovascular side effects driven by microbiota and inflammation.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Diet , Gastrointestinal Microbiome , Homeostasis , Interleukin-23/metabolism , Interleukins/metabolism , Animals , Atherosclerosis/pathology , Biomarkers , Disease Models, Animal , Disease Progression , Gene Expression , Immunophenotyping , Interleukin-23/deficiency , Lipid Metabolism , Mice , Mice, Knockout , Osteopontin/genetics , Osteopontin/metabolism , Signal Transduction , Interleukin-22
9.
Sci Rep ; 7(1): 2255, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28536468

ABSTRACT

Myeloid cells, key players in atherosclerosis, take up and present antigens, leading to systemic and local T cell activation. The recruitment and activation of immune cells to the aorta in atherosclerosis is regulated by adhesion molecules, chemokines and cytokines. IL-27R is an immunoregulatory signaling nod in autoimmune and infectious pathologies. IL-27R was shown to suppress T cells activation in atherosclerosis, however it's possible role in myeloid cell accumulation and activation is not understood. Here we demonstrate that Apoe -/- Il27ra -/- mice fed with "Western Diet" for 7 or 18 weeks developed significantly more atherosclerosis compared to Apoe -/- Il27ra +/- controls. Accelerated disease was driven by enhanced expression of adhesion molecules and chemokines causing the accumulation of immune cells. Myeloid cells produced more inflammatory cytokines and upregulated MHCII. Multiphoton microscopy revealed more efficient interactions between aortic myeloid cells and CD4+ T cells. Overall, we show that IL-27R signaling controls endothelial cells activation and myeloid cell recruitment at early and advanced stages of atherosclerosis. In the absence of IL-27R myeloid cells become hyperactivated, produce pro-inflammatory cytokines and act as more potent antigen presenting cells. Enhanced interactions between Il27ra -/- APC and CD4+ T cells in the aortic wall contribute to T cells re-activation and pro-atherogenic cytokine production.


Subject(s)
Antigen Presentation/immunology , Atherosclerosis/etiology , Atherosclerosis/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Atherosclerosis/pathology , Biomarkers , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression , Immunophenotyping , Male , Mice , Mice, Knockout , Receptors, Interleukin/genetics
10.
FEBS J ; 283(9): 1636-52, 2016 05.
Article in English | MEDLINE | ID: mdl-26700480

ABSTRACT

Cardiovascular diseases (CVD) are the major cause of death in developed countries. Various risk factors including host genetics and, more importantly, environmental factors such as lifestyle, diet and smoking influence CVD progression. Two common forms of CVD are atherosclerosis and abdominal aortic aneurysm (AAA). Emerging evidence suggests that inflammation plays a pivotal role in CVD. However, it remains unclear whether the same inflammatory pathways prove essential for atherosclerosis and AAA because, in some cases, the same mechanisms uniformly promote both diseases, while in others they function in opposite ways. Cytokines, key mediators of inflammation, play an important role in the development of atherosclerosis but have only been scarcely studied in AAA. In this review, we discuss the importance of immune-mediated mechanisms and cytokines in the pathogenesis of atherosclerosis and AAA.


Subject(s)
Angiotensin II/genetics , Aortic Aneurysm, Abdominal/genetics , Atherosclerosis/genetics , Cytokines/genetics , Angiotensin II/immunology , Animals , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cytokines/immunology , Diet, Western/adverse effects , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammation , Risk Factors , Sedentary Behavior , Smoking/adverse effects , Smoking/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...