Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Curr Opin Immunol ; 72: 286-297, 2021 10.
Article in English | MEDLINE | ID: covidwho-1606955

ABSTRACT

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is caused by mutations in the Autoimmune Regulator (AIRE) gene, which impair the thymic negative selection of self-reactive T-cells and underlie the development of autoimmunity that targets multiple endocrine and non-endocrine tissues. Beyond autoimmunity, APECED features heightened susceptibility to certain specific infections, which is mediated by anti-cytokine autoantibodies and/or T-cell driven autoimmune tissue injury. These include the 'signature' APECED infection chronic mucocutaneous candidiasis (CMC), but also life-threatening coronavirus disease 2019 (COVID-19) pneumonia, bronchiectasis-associated bacterial pneumonia, and sepsis by encapsulated bacteria. Here we discuss the expanding understanding of the immunological mechanisms that contribute to infection susceptibility in this prototypic syndrome of impaired central tolerance, which provide the foundation for devising improved diagnostic and therapeutic strategies for affected patients.


Subject(s)
COVID-19/immunology , Candidiasis, Cutaneous/immunology , Polyendocrinopathies, Autoimmune/immunology , T-Lymphocytes/immunology , Transcription Factors/genetics , Animals , Autoimmunity , Bronchiectasis , COVID-19/epidemiology , COVID-19/genetics , Candidiasis, Cutaneous/epidemiology , Candidiasis, Cutaneous/genetics , Clonal Selection, Antigen-Mediated/genetics , Disease Susceptibility , Humans , Immune Tolerance/genetics , Polyendocrinopathies, Autoimmune/epidemiology , Polyendocrinopathies, Autoimmune/genetics
2.
Front Immunol ; 12: 755579, 2021.
Article in English | MEDLINE | ID: covidwho-1556334

ABSTRACT

During the COVID-19 pandemic, a phenomenon emerged in which some patients with severe disease were critically ill and could not be discharged from the ICU even though they exhibited negative viral tests. To explore the underlying mechanism, we collected blood samples from these patients and analyzed the gene expression profiles of peripheral immune cells. We found that all enrolled patients, regardless of changes in genes related to different symptoms and inflammatory responses, showed universally and severely decreased expression of adaptive immunity-related genes, especially those related to T/B cell arms and HLA molecules, and that these patients exhibited long-term secondary infections. In addition, no significant change was found in the expression of classic immunosuppression molecules including PD-1, PD-L1, and CTLA-4, suggesting that the adaptive immune suppression may not be due to the change of these genes. According to the published literatures and our data, this adaptive immunosuppression is likely to be caused by the "dysregulated host response" to severe infection, similar to the immunosuppression that exists in other severely infected patients with sepsis.


Subject(s)
Adaptive Immunity/immunology , COVID-19/immunology , Immune Tolerance/immunology , Adaptive Immunity/genetics , Aged , COVID-19/diagnosis , COVID-19/genetics , Coinfection/diagnosis , Coinfection/genetics , Coinfection/immunology , Cross-Sectional Studies , Cytokine Release Syndrome/genetics , Female , Gene Expression Profiling , Humans , Immune Tolerance/genetics , Inflammation/genetics , Intensive Care Units , Male , Middle Aged , Patient Discharge , SARS-CoV-2/isolation & purification , Smell/genetics , Taste/genetics
4.
Cytometry A ; 97(12): 1217-1221, 2020 12.
Article in English | MEDLINE | ID: covidwho-893217

ABSTRACT

Several months after the sudden emergence of SARS-CoV-2 and COVID-19, the understanding of the appropriate host immune response to a virus totally unknown of human immune surveillance is still of major importance. By international definition, COVID-19 falls in the scope of septic syndromes (organ dysfunction due to dysregulated host response to an infection) in which immunosuppression is a significant driver of mortality. Sepsis-induced immunosuppression is mostly defined and monitored by the measurement of decreased expression of HLA-DR molecules on circulating monocytes (mHLA-DR). In this interim review, we summarize the first mHLA-DR results in COVID-19 patients. In critically ill patients, results homogenously indicate a decreased mHLA-DR expression, which, along with profound lymphopenia and other functional alterations, is indicative of a status of immunosuppression. © 2020 International Society for Advancement of Cytometry.


Subject(s)
COVID-19/immunology , HLA-DR Antigens/immunology , Monocytes/immunology , COVID-19/pathology , COVID-19/virology , Female , Flow Cytometry , HLA-DR Antigens/genetics , Humans , Immune Tolerance/genetics , Male , SARS-CoV-2/pathogenicity
5.
Immunol Cell Biol ; 99(2): 177-191, 2021 02.
Article in English | MEDLINE | ID: covidwho-894767

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a zoonosis like most of the great plagues sculpting human history, from smallpox to pandemic influenza and human immunodeficiency virus. When viruses jump into a new species the outcome of infection ranges from asymptomatic to lethal, historically ascribed to "genetic resistance to viral disease." People have exploited these differences for good and bad, for developing vaccines from cowpox and horsepox virus, controlling rabbit plagues with myxoma virus and introducing smallpox during colonization of America and Australia. Differences in resistance to viral disease are at the core of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) crisis, yet our understanding of the mechanisms in any interspecies leap falls short of the mark. Here I review how the two key parameters of viral disease are countered by fundamentally different genetic mechanisms for resistance: (1) virus transmission, countered primarily by activation of innate and adaptive immune responses; and (2) pathology, countered primarily by tolerance checkpoints to limit innate and adaptive immune responses. I discuss tolerance thresholds and the role of CD8 T cells to limit pathological immune responses, the problems posed by tolerant superspreaders and the signature coronavirus evasion strategy of eliciting only short-lived neutralizing antibody responses. Pinpointing and targeting the mechanisms responsible for varying pathology and short-lived antibody were beyond reach in previous zoonoses, but this time we are armed with genomic technologies and more knowledge of immune checkpoint genes. These known unknowns must now be tackled to solve the current COVID-19 crisis and the inevitable zoonoses to follow.


Subject(s)
COVID-19 , Immune Tolerance/immunology , Immunity, Innate/genetics , SARS-CoV-2/immunology , Virus Diseases/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/physiology , Antibodies, Viral/immunology , COVID-19/genetics , COVID-19/immunology , COVID-19/virology , Humans , Immune Tolerance/genetics , Rabbits , SARS-CoV-2/genetics , Viral Zoonoses/genetics , Viral Zoonoses/immunology , Virus Diseases/genetics
6.
FEBS J ; 288(17): 5055-5070, 2021 09.
Article in English | MEDLINE | ID: covidwho-894751

ABSTRACT

The ongoing coronavirus disease 2019 (COVID-19) crisis caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has triggered a large-scale pandemic that is afflicting millions of individuals in over 200 countries. The clinical spectrum caused by SARS-CoV-2 infections can range from asymptomatic infection to mild undifferentiated febrile illness to severe respiratory disease with multiple complications. Elderly patients (aged 60 and above) with comorbidities such as cardiovascular diseases and diabetes mellitus appear to be at highest risk of a severe disease outcome. To protect against pulmonary immunopathology caused by SARS-CoV-2 infection, the host primarily depends on two distinct defense strategies: resistance and disease tolerance. Resistance is the ability of the host to suppress and eliminate incoming viruses. By contrast, disease tolerance refers to host responses that promote host health regardless of their impact on viral replication. Disruption of either resistance or disease tolerance mechanisms or both could underpin predisposition to elevated risk of severe disease during viral infection. Aging can disrupt host resistance and disease tolerance by compromising immune functions, weakening of the unfolded protein response, progressive mitochondrial dysfunction, and altering metabolic processes. A comprehensive understanding of the molecular mechanisms underlying declining host defense in elderly individuals could thus pave the way to provide new opportunities and approaches for the treatment of severe COVID-19.


Subject(s)
Aging/genetics , COVID-19/genetics , Disease Resistance/genetics , SARS-CoV-2/pathogenicity , Aged , COVID-19/pathology , COVID-19/virology , Host-Pathogen Interactions/genetics , Humans , Immune Tolerance/genetics , Pandemics , SARS-CoV-2/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL