Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Immunother Cancer ; 11(5)2023 05.
Article in English | MEDLINE | ID: covidwho-20233460

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer. However, only a portion of patients respond to such treatments. Therefore, it remains a prevailing clinical need to identify factors associated with acquired resistance or lack of response to ICIs. We hypothesized that the immunosuppressive CD71+ erythroid cells (CECs) within the tumor and/or distant 'out-of-field' may impair antitumor response. METHODS: We studied 38 patients with cancer through a phase II clinical trial investigating the effects of oral valproate combined with avelumab (anti-programmed death-ligand 1 (PD-L1)) in virus-associated solid tumors (VASTs). We quantified the frequency/functionality of CECs in blood and biopsies of patients. Also, we established an animal model of melanoma (B16-F10) to investigate the possible effects of erythropoietin (EPO) treatment on anti-PD-L1 therapy. RESULTS: We found a substantial expansion of CECs in the blood of patients with VAST compared with healthy controls. We noted that the frequency of CECs in circulation was significantly higher at the baseline and throughout the study in non-responders versus responders to PD-L1 therapy. Moreover, we observed that CECs in a dose-dependent manner suppress effector functions of autologous T cells in vitro. The subpopulation of CD45+CECs appears to have a more robust immunosuppressive property compared with their CD45- counterparts. This was illustrated by a stronger expression of reactive oxygen species, PD-L1/PD-L2, and V-domain Ig suppressor of T-cell activation in this subpopulation. Lastly, we found a higher frequency of CECs in the blood circulation at the later cancer stage and their abundance was associated with anemia, and a poor response to immunotherapy. Finally, we report the expansion of CECs in the spleen and tumor microenvironment of mice with melanoma. We found that although CECs in tumor-bearing mice secret artemin, this was not the case for VAST-derived CECs in humans. Notably, our results imply that EPO, a frequently used drug for anemia treatment in patients with cancer, may promote the generation of CECs and subsequently abrogates the therapeutic effects of ICIs (eg, anti-PD-L1). CONCLUSIONS: Our results demonstrate that anemia by the expansion of CECs may enhance cancer progression. Notably, measuring the frequency of CECs may serve as a valuable biomarker to predict immunotherapy outcomes.


Subject(s)
Melanoma , T-Lymphocytes , Humans , Animals , Mice , T-Lymphocytes/pathology , Immunotherapy/methods , Erythroid Cells/pathology , Neoplasm Staging , Tumor Microenvironment
3.
Gynakologie ; 55(9):654-659, 2022.
Article in German | EMBASE | ID: covidwho-2283384

ABSTRACT

Vaccinations are one of the greatest achievements for protecting public health. Vaccines given to pregnant women protect not only the pregnant woman, but also the newborn. Pregnant women are disproportionately strongly affected by infections. The conflicting demands on the maternal immune system during pregnancy geared toward maintaining fetal immune tolerance make a rapid and effective immune response against pathogens more difficult. This dynamic state of immune adaptation predisposes pregnant women to more severe disease progression. Vaccination can prevent infection or a serious course of disease. As a result, the risk of premature birth and other serious pregnancy complications that can have lifelong consequences for both mother and child also decreases. After birth, when the newborn must first develop an adaptive memory for a hitherto unknown, antigen-rich environment, it is particularly vulnerable to infections and resulting complications. The transfer of maternal antibodies across the placenta protects infants who are too young to be vaccinated. When breastfeeding, this continues through antibodies in breast milk. For the vaccinations recommended by the Standing Vaccination Committee (STIKO) during pregnancy (influenza, pertussis, coronavirus disease [COVID]-19), there is clear evidence from various observational and prospective studies that they protect mother and child either from infection or from a severe disease course. The following article gives an overview of the vaccination strategy for pregnancy and summarizes the scientific data on effectiveness of the vaccinations currently recommended during pregnancy.Copyright © 2022, The Author(s), under exclusive licence to Springer Medizin Verlag GmbH, ein Teil von Springer Nature.

4.
Microbiol Spectr ; 11(3): e0429222, 2023 Jun 15.
Article in English | MEDLINE | ID: covidwho-2271377

ABSTRACT

Globally, COVID-19 vaccines have emerged as a boon, especially during the severe pandemic phases to control the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, saving millions of lives. However, mixed responses to vaccination with breakthrough challenges provided a rationale to explore the immune responses generated postvaccination, which plausibly alter the subsequent course of infection. In this regard, we comprehensively profiled the nasopharyngeal transcriptomic signature of double-dose-vaccinated individuals with breakthrough infections in comparison to unvaccinated infected persons. The vaccinated individuals demonstrated a gross downregulation of ribosomal proteins along with immune response genes and transcription/translational machinery that methodically modulated the entire innate immune landscape toward immune tolerance, a feature of innate immune memory. This coordinated response was orchestrated through 17 transcription factors captured as differentially expressed in the vaccination breakthroughs, including epigenetic modulators of CHD1 and LMNB1 and several immune response effectors, with ELF1 emerging as one of the important transcriptional regulators of the antiviral innate immune response. Deconvolution algorithm using bulk gene expression data revealed decreased T-cell populations with higher expression of memory B cells in the vaccination breakthroughs. Thus, vaccination might synergize the innate immune response with humoral and T-cell correlates of protection to more rapidly clear SARS-CoV-2 infections and reduce symptoms within a shorter span of time. An important feature invariably noted after secondary vaccination is downregulation of ribosomal proteins, which might plausibly be an important factor arising from epigenetic reprogramming leading to innate immune tolerance. IMPORTANCE The development of multiple vaccines against SARS-CoV-2 infection is an unprecedented milestone achieved globally. Immunization of the mass population is a rigorous process for getting the pandemic under control, yet continuous challenges are being faced, one of them being breakthrough infections. This is the first study wherein the vaccination breakthrough cases of COVD-19 relative to unvaccinated infected individuals have been explored. In the context of vaccination, how do innate and adaptive immune responses correspond to SARS-CoV-2 infection? How do these responses culminate in a milder observable phenotype with shorter hospital stay in vaccination breakthrough cases compared with the unvaccinated? We identified a subdued transcriptional landscape in vaccination breakthroughs with decreased expression of a large set of immune and ribosomal proteins genes. We propose a module of innate immune memory, i.e., immune tolerance, which plausibly helps to explain the observed mild phenotype and fast recovery in vaccination breakthroughs.


Subject(s)
COVID-19 Vaccines , COVID-19 , Humans , SARS-CoV-2/genetics , COVID-19/prevention & control , Vaccination , Immunity, Innate , Breakthrough Infections
5.
Die Gynäkologie ; 55(9):654-659, 2022.
Article in German | Web of Science | ID: covidwho-2014084

ABSTRACT

Vaccinations are one of the greatest achievements for protecting public health. Vaccines given to pregnant women protect not only the pregnant woman, but also the newborn. Pregnant women are disproportionately strongly affected by infections. The conflicting demands on the maternal immune system during pregnancy geared toward maintaining fetal immune tolerance make a rapid and effective immune response against pathogens more difficult. This dynamic state of immune adaptation predisposes pregnant women to more severe disease progression. Vaccination can prevent infection or a serious course of disease. As a result, the risk of premature birth and other serious pregnancy complications that can have lifelong consequences for both mother and child also decreases. After birth, when the newborn must first develop an adaptive memory for a hitherto unknown, antigen-rich environment, it is particularly vulnerable to infections and resulting complications. The transfer of maternal antibodies across the placenta protects infants who are too young to be vaccinated. When breastfeeding, this continues through antibodies in breast milk. For the vaccinations recommended by the Standing Vaccination Committee (STIKO) during pregnancy (influenza, pertussis, coronavirus disease [COVID]-19), there is clear evidence from various observational and prospective studies that they protect mother and child either from infection or from a severe disease course. The following article gives an overview of the vaccination strategy for pregnancy and summarizes the scientific data on effectiveness of the vaccinations currently recommended during pregnancy.

6.
Front Cell Neurosci ; 16: 918883, 2022.
Article in English | MEDLINE | ID: covidwho-1911070

ABSTRACT

Prion diseases such as Creutzfeldt-Jakob disease in humans, bovine spongiform encephalopathy in cattle, and scrapie in sheep, are infectious and chronic neurodegenerative diseases to which there are no cures. Infection with prions in the central nervous system (CNS) ultimately causes extensive neurodegeneration, and this is accompanied by prominent microglial and astrocytic activation in affected regions. The microglia are the CNS macrophages and help maintain neuronal homeostasis, clear dead or dying cells and provide defense against pathogens. The microglia also provide neuroprotection during CNS prion disease, but their pro-inflammatory activation may exacerbate the development of the neuropathology. Innate immune tolerance induced by consecutive systemic bacterial lipopolysaccharide (LPS) treatment can induce long-term epigenetic changes in the microglia in the brain that several months later can dampen their responsiveness to subsequent LPS treatment and impede the development of neuritic damage in a transgenic mouse model of Alzheimer's disease-like pathology. We therefore reasoned that innate immune tolerance in microglia might similarly impede the subsequent development of CNS prion disease. To test this hypothesis groups of mice were first infected with prions by intracerebral injection, and 35 days later given four consecutive systemic injections with LPS to induce innate immune tolerance. Our data show that consecutive systemic LPS treatment did not affect the subsequent development of CNS prion disease. Our data suggests innate immune tolerance in microglia does not influence the subsequent onset of prion disease-induced neuropathology in mice, despite previously published evidence of this effect in an Alzheimer's disease mouse model.

7.
J Leukoc Biol ; 111(2): 497-508, 2022 02.
Article in English | MEDLINE | ID: covidwho-1669515

ABSTRACT

Coronaviruses (CoVs) are RNA viruses that cause human respiratory infections. Zoonotic transmission of the SARS-CoV-2 virus caused the recent COVID-19 pandemic, which led to over 2 million deaths worldwide. Elevated inflammatory responses and cytotoxicity in the lungs are associated with COVID-19 severity in SARS-CoV-2-infected individuals. Bats, which host pathogenic CoVs, operate dampened inflammatory responses and show tolerance to these viruses with mild clinical symptoms. Delineating the mechanisms governing these host-specific inflammatory responses is essential to understand host-virus interactions determining the outcome of pathogenic CoV infections. Here, we describe the essential role of inflammasome activation in determining COVID-19 severity in humans and innate immune tolerance in bats that host several pathogenic CoVs. We further discuss mechanisms leading to inflammasome activation in human SARS-CoV-2 infection and how bats are molecularly adapted to suppress these inflammasome responses. We also report an analysis of functionally important residues of inflammasome components that provide new clues of bat strategies to suppress inflammasome signaling and innate immune responses. As spillover of bat viruses may cause the emergence of new human disease outbreaks, the inflammasome regulation in bats and humans likely provides specific strategies to combat the pathogenic CoV infections.


Subject(s)
COVID-19/pathology , Immune Tolerance , Immunity, Innate , Inflammasomes/immunology , SARS-CoV-2/immunology , Animals , COVID-19/immunology , COVID-19/virology , Chiroptera , Humans , Inflammasomes/metabolism , Phylogeny
8.
Trends Immunol ; 43(2): 148-162, 2022 02.
Article in English | MEDLINE | ID: covidwho-1634995

ABSTRACT

Ubc13-catalyzed K63 ubiquitination is a major control point for immune signaling. Recent evidence has shown that the control of multiple immune functions, including chronic inflammation, pathogen responses, lymphocyte activation, and regulatory signaling, is altered by K63 ubiquitination. In this review, we detail the novel cellular sensors that are dependent on K63 ubiquitination for their function in the immune signaling network. Many pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can target K63 ubiquitination to inhibit pathogen immune responses; we describe novel details of the pathways involved and summarize recent clinically relevant SARS-CoV-2-specific responses. We also discuss recent evidence that regulatory T cell (Treg) versus T helper (TH) 1 and TH17 cell subset regulation might involve K63 ubiquitination. Knowledge gaps that merit future investigation and clinically relevant pathways are also addressed.


Subject(s)
COVID-19 , Lysine , Humans , Lysine/metabolism , SARS-CoV-2 , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Life Sci ; 286: 120063, 2021 Dec 01.
Article in English | MEDLINE | ID: covidwho-1472089

ABSTRACT

COVID-19 is a multi-faceted disease ranging from asymptomatic to severely ill condition that primarily affects the lungs and could advance to other organs as well. It's causing factor, SARS-CoV-2 is recognized to develop robust cell-mediated immunity that responsible to either control or exaggerate the infection. As an important cell subset that control immune responses and are significantly dysregulated in COVID-19, Tregs is proposed to be considered for COVID-19 management. Among its hallmark, TNFR2 is recently recognized to play important role in the function and survival of Tregs. This review gathers available TNFR2 agonists to directly target Tregs as a potential approach to overcome immune dysregulation that affect the severity in COVID-19. Furthermore, this review performs a rigid body docking of TNF-TNFR2 interaction and such interaction with TNFR2 agonist to predict the optimal targeting approach.


Subject(s)
COVID-19/immunology , COVID-19/therapy , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/immunology , COVID-19/virology , Humans , Lymphocyte Activation , SARS-CoV-2/isolation & purification
10.
J Pers Med ; 11(8)2021 Jul 27.
Article in English | MEDLINE | ID: covidwho-1376868

ABSTRACT

A 57-year female patient diagnosed with Behçet's disease, on azathioprine, was noticed to have at a routine examination antinuclear and antiphospholipid antibodies. An overlapping lupus-like syndrome was diagnosed; hydroxychloroquine and aspirin were added. Three years later, the patient presented with dyspnea and sweating, with no fever. A cardiac bruit was noted; a giant vegetation was detected by echocardiography. Laboratory revealed severe thrombocytopenia, antiphospholipid antibodies and low complement. Blood cultures were positive for Abiotrophia defectiva serology and also revealed a chronic Coxiella burnetii infection. Antibiotic therapy, low-dose anticoagulation and control of the underlying disease mildly improved the platelet count, which fully recovered only after cardiac valve replacement. However, the Behçet's disease, initially quiescent, flared after the therapy of infections. We discuss potential links between Behçet's disease and the occurrence of antinuclear and antiphospholipid antibodies and Coxiella endocarditis in this setting. We also highlight the differences between the endocarditis in Behçet's disease, antiphospholipid syndrome, Coxiella burnetii and Abiotrophia defectiva infection, respectively. Intracellular infections may modify the presentation of autoimmune diseases. Confounding clinical features of Coxiella persistent infection and non-bacterial thrombotic endocarditis in Behçet's disease warrant further insight.

11.
Front Immunol ; 12: 659621, 2021.
Article in English | MEDLINE | ID: covidwho-1285289

ABSTRACT

Methods for suppressing the host immune system over the long term and improving transplantation tolerance remain a primary issue in organ transplantation. Cell therapy is an emerging therapeutic strategy for immunomodulation after transplantation. Mesenchymal stem cells (MSCs) are adult multipotent stem cells with wide differentiation potential and immunosuppressive properties, which are mostly used in regenerative medicine and immunomodulation. In addition, emerging research suggests that MSC-derived exosomes have the same therapeutic effects as MSCs in many diseases, while avoiding many of the risks associated with cell transplantation. Their unique immunomodulatory properties are particularly important in the immune system-overactive graft environment. In this paper, we review the effects of MSC-derived exosomes in the immune regulation mechanism after organ transplantation and graft-versus-host disease (GvHD) from various perspectives, including immunosuppression, influencing factors, anti-inflammatory properties, mediation of tissue repair and regeneration, and the induction of immune tolerance. At present, the great potential of MSC-derived exosomes in immunotherapy has attracted a great deal of attention. Furthermore, we discuss the latest insights on MSC-derived exosomes in organ transplantation and GvHD, especially its commercial production concepts, which aim to provide new strategies for improving the prognosis of organ transplantation patients.


Subject(s)
Exosomes/immunology , Immunomodulation/immunology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Organ Transplantation/methods , Transplantation Tolerance/immunology , Adult , Exosomes/metabolism , Graft vs Host Disease/immunology , Humans , Mesenchymal Stem Cells/metabolism , Regenerative Medicine/methods
12.
Proc Jpn Acad Ser B Phys Biol Sci ; 97(6): 277-291, 2021.
Article in English | MEDLINE | ID: covidwho-1267427

ABSTRACT

Cytokines are important intercellular communication tools for immunity. Most cytokines utilize the JAK-STAT and Ras-ERK pathways to promote gene transcription and proliferation; however, this signaling is tightly regulated. The suppressor of cytokine signaling (SOCS) family and SPRED family are a representative negative regulators of the JAK-STAT pathway and the Ras-ERK pathway, respectively. The SOCS family regulates the differentiation and function of CD4+ T cells, CD8+ T cells, and regulatory T cells, and is involved in immune tolerance, anergy, and exhaustion. SPRED family proteins have been shown to inactivate Ras by recruiting the Ras-GTPase neurofibromatosis type 1 (NF1) protein. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergies, and tumorigenesis, and SPRED1 is involved in NF1-like syndromes and tumors. We also identified the NR4a family of nuclear receptors as a key transcription factor for immune tolerance that suppresses cytokine expression and induces various immuno-regulatory molecules including SOCS1.


Subject(s)
CD8-Positive T-Lymphocytes , Suppressor of Cytokine Signaling Proteins , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Humans , Immune Tolerance , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism
13.
Cell Immunol ; 362: 104302, 2021 04.
Article in English | MEDLINE | ID: covidwho-1085577

ABSTRACT

MDSC are a heterogeneous population of immature myeloid cells that are released by biological stress such as tissue damage and inflammation. Conventionally, MDSC are known for their detrimental role in chronic inflammation and neoplastic conditions. However, their intrinsic functions in immunoregulation, wound healing, and angiogenesis are intended to protect from over-reactive immune responses, maintenance of immunotolerance, tissue repair, and homeostasis. Paradoxically, under certain conditions, MDSC can impair protective immune responses and exacerbate the disease. The transition from protective to harmful MDSC is most likely driven by environmental and epigenetic mechanisms induced by prolonged exposure to unresolved inflammatory triggers. Here, we review several examples of the dual impact of MDSC in conditions such as maternal-fetal tolerance, self-antigens immunotolerance, obesity-associated cancer, sepsis and trauma. Moreover, we also highlighted the evidence indicating that MDSC have a role in COVID-19 pathophysiology. Finally, we have summarized the evidence indicating epigenetic mechanisms associated with MDSC function.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Animals , COVID-19/immunology , Epigenesis, Genetic , Female , Humans , Immune Tolerance/immunology , Inflammation/immunology , Male , Neoplasms/immunology , Obesity/immunology , Pregnancy , Wound Healing/immunology
SELECTION OF CITATIONS
SEARCH DETAIL