Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell Calcium ; 101: 102524, 2022 01.
Article in English | MEDLINE | ID: covidwho-1914206

ABSTRACT

A recent publication proposes that T cell receptor activation elicits formation of the Ca2+ releasing messenger NAADP from NAADPH, catalysed by the NADPH oxidase DUOX. This is in contrast to the hitherto prevailing view that CD38 is critical for NAADP formation. Is it time to reassess the role of CD38?


Subject(s)
Calcium , Membrane Glycoproteins , ADP-ribosyl Cyclase 1/metabolism , Calcium/metabolism , Calcium Signaling , Membrane Glycoproteins/metabolism , NADP/metabolism , Receptors, Antigen, T-Cell
3.
Biochem J ; 478(23): 4071-4092, 2021 12 10.
Article in English | MEDLINE | ID: covidwho-1556088

ABSTRACT

The COVID-19 pandemic reminds us that in spite of the scientific progress in the past century, there is a lack of general antiviral strategies. In analogy to broad-spectrum antibiotics as antibacterial agents, developing broad spectrum antiviral agents would buy us time for the development of vaccines and treatments for future viral infections. In addition to targeting viral factors, a possible strategy is to understand host immune defense mechanisms and develop methods to boost the antiviral immune response. Here we summarize the role of NAD+-consuming enzymes in the immune defense against viral infections, with the hope that a better understanding of this process could help to develop better antiviral therapeutics targeting these enzymes. These NAD+-consuming enzymes include PARPs, sirtuins, CD38, and SARM1. Among these, the antiviral function of PARPs is particularly important and will be a focus of this review. Interestingly, NAD+ biosynthetic enzymes are also implicated in immune responses. In addition, many viruses, including SARS-CoV-2 contain a macrodomain-containing protein (NSP3 in SARS-CoV-2), which serves to counteract the antiviral function of host PARPs. Therefore, NAD+ and NAD+-consuming enzymes play crucial roles in immune responses against viral infections and detailed mechanistic understandings in the future will likely facilitate the development of general antiviral strategies.


Subject(s)
Antiviral Agents/therapeutic use , Immunity, Innate , NAD/metabolism , Virus Diseases/drug therapy , ADP-ribosyl Cyclase 1/metabolism , Armadillo Domain Proteins/metabolism , COVID-19/immunology , Cytoskeletal Proteins/metabolism , Humans , NAD/immunology , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Domains , SARS-CoV-2 , Sirtuins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Diseases/immunology , COVID-19 Drug Treatment
4.
J Allergy Clin Immunol ; 149(3): 912-922, 2022 03.
Article in English | MEDLINE | ID: covidwho-1536619

ABSTRACT

BACKGROUND: Multisystem inflammatory syndrome in children (MIS-C) is an acute, febrile, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-associated syndrome, often with cardiohemodynamic dysfunction. Insight into mechanism of disease is still incomplete. OBJECTIVE: Our objective was to analyze immunologic features of MIS-C patients compared to febrile controls (FC). METHODS: MIS-C patients were defined by narrow criteria, including having evidence of cardiohemodynamic involvement and no macrophage activation syndrome. Samples were collected from 8 completely treatment-naive patients with MIS-C (SARS-CoV-2 serology positive), 3 patients with unclassified MIS-C-like disease (serology negative), 14 FC, and 5 MIS-C recovery (RCV). Three healthy controls (HCs) were used for comparisons of normal range. Using spectral flow cytometry, we assessed 36 parameters in antigen-presenting cells (APCs) and 29 in T cells. We used biaxial analysis and uniform manifold approximation and projection (UMAP). RESULTS: Significant elevations in cytokines including CXCL9, M-CSF, and IL-27 were found in MIS-C compared to FC. Classic monocytes and type 2 dendritic cells (DCs) were downregulated (decreased CD86, HLA-DR) versus HCs; however, type 1 DCs (CD11c+CD141+CLEC9A+) were highly activated in MIS-C patients versus FC, expressing higher levels of CD86, CD275, and atypical conventional DC markers such as CD64, CD115, and CX3CR1. CD169 and CD38 were upregulated in multiple monocyte subtypes. CD56dim/CD57-/KLRGhi/CD161+/CD38- natural killer (NK) cells were a unique subset in MIS-C versus FC without macrophage activation syndrome. CONCLUSION: Orchestrated by complex cytokine signaling, type 1 DC activation and NK dysregulation are key features in the pathophysiology of MIS-C. NK cell findings may suggest a relationship with macrophage activation syndrome, while type 1 DC upregulation implies a role for antigen cross-presentation.


Subject(s)
COVID-19/complications , Dendritic Cells/immunology , Dendritic Cells/virology , SARS-CoV-2/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/virology , ADP-ribosyl Cyclase 1/blood , Adolescent , Antigens, Viral/immunology , COVID-19/immunology , COVID-19/virology , Case-Control Studies , Child , Child, Preschool , Cross-Priming , Cytokines/blood , Dendritic Cells/classification , Female , HLA-DR Antigens/blood , Humans , Immunophenotyping , Interferon-gamma/blood , Interleukins/blood , Killer Cells, Natural/immunology , Male , Membrane Glycoproteins/blood , Models, Immunological , Monocytes/immunology , Sialic Acid Binding Ig-like Lectin 1/blood , T-Lymphocytes/immunology , T-Lymphocytes/virology , Up-Regulation
5.
J Clin Lab Anal ; 35(11): e24046, 2021 Nov.
Article in English | MEDLINE | ID: covidwho-1525449

ABSTRACT

BACKGROUND: Few studies have investigated the alterations in the T and B cell counts and related subgroups in pulmonary infections especially COVID-19. Here, we aimed to evaluate total T and B lymphocytes and T cell subgroup counts to find the possible correlation between number of these cells and severity and mortality in COVID-19 patients. METHODS: This study was performed on 40 patients with severe COVID-19 infection confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and chest HRCT in August 2020. By the time of admission, T lymphocytes profile in peripheral blood was investigated using multicolor flow cytometry. The total number of T lymphocytes, CD4+ T cells, CD8+ T cells, and B lymphocytes were calculated. Expression of CD2, CD3, CD5, and CD7 as pan T cell surface markers and expression of CD38 and HLA-DR as activated markers on T lymphocytes were also evaluated. RESULTS: Nine patients (22.5%) died during the study and 16 patients (40%) were admitted to ICU. Deceased patients demonstrated lower amounts of T cell count and CD4+ T cell count (with a marginal difference (p = 0.07)) compared with survived patients at the time of admission. The chance of mortality was significantly higher for patients with CD7 loss (OR = 14.89). A marginally significant relationship was also indicated between CD4<200/ml and mortality (OR = 8.65), but no other significant relationships were observed between variables and ICU admission. CONCLUSION: Altogether, CD7 loss on T lymphocytes and CD4+ T cell count below 200/ml revealed a significant relationship with mortality. Considering T lymphocytes and T cell subgroup count could have a predictive value for patients suffering from COVID-19.


Subject(s)
COVID-19/immunology , Lymphocyte Subsets , SARS-CoV-2 , ADP-ribosyl Cyclase 1/analysis , Antigens, CD7/analysis , COVID-19/mortality , Female , Humans , Male , Middle Aged , Severity of Illness Index
6.
Physiol Rev ; 101(4): 1457-1486, 2021 10 01.
Article in English | MEDLINE | ID: covidwho-1443666

ABSTRACT

This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti-viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , COVID-19/metabolism , Membrane Glycoproteins/metabolism , SARS-CoV-2 , ADP-ribosyl Cyclase 1/genetics , COVID-19/pathology , COVID-19/virology , Gene Expression Regulation, Enzymologic , Humans , Membrane Glycoproteins/genetics
7.
Viruses ; 13(10)2021 09 30.
Article in English | MEDLINE | ID: covidwho-1444330

ABSTRACT

BACKGROUND: The immunological changes associated with COVID-19 are largely unknown. METHODS: Patients with COVID-19 showing moderate (n = 18; SpO2 > 93%, respiratory rate > 22 per minute, CRP > 10 mg/L) and severe (n = 23; SpO2 < 93%, respiratory rate >30 per minute, PaO2/FiO2 ≤ 300 mmHg, permanent oxygen therapy, qSOFA > 2) infection, and 37 healthy donors (HD) were enrolled. Circulating T- and B-cell subsets were analyzed by flow cytometry. RESULTS: CD4+Th cells were skewed toward Th2-like phenotypes within CD45RA+CD62L- (CM) and CD45RA-CD62L- (EM) cells in patients with severe COVID-19, while CM CCR6+ Th17-like cells were decreased if compared with HD. Within CM Th17-like cells "classical" Th17-like cells were increased and Th17.1-like cells were decreased in severe COVID-19 cases. Circulating CM follicular Th-like (Tfh) cells were decreased in all COVID-19 patients, and Tfh17-like cells represented the most predominant subset in severe COVID-19 cases. Both groups of patients showed increased levels of IgD-CD38++ B cells, while the levels of IgD+CD38- and IgD-CD38- were decreased. The frequency of IgD+CD27+ and IgD-CD27+ B cells was significantly reduced in severe COVID-19 cases. CONCLUSIONS: We showed an imbalance within almost all circulating memory Th subsets during acute COVID-19 and showed that altered Tfh polarization led to a dysregulated humoral immune response.


Subject(s)
B-Lymphocyte Subsets/immunology , COVID-19/immunology , Immunity , SARS-CoV-2 , ADP-ribosyl Cyclase 1 , Adult , Aged , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Humans , Immunoglobulin D , Male , Middle Aged , Oxygen , Receptors, CCR6 , T-Lymphocytes/metabolism , Th17 Cells/immunology
8.
Physiol Rev ; 102(1): 339-341, 2022 01 01.
Article in English | MEDLINE | ID: covidwho-1398740

ABSTRACT

During the COVID-19 pandemic, efforts have been made worldwide to develop effective therapies to address the devastating immune-mediated effects of SARS-CoV-2. With the exception of monoclonal antibody-mediated therapeutics and preventive approaches such as mass immunization, most experimental or repurposed drugs have failed in large randomized clinical trials (https://www.who.int/publications/i/item/therapeutics-and-covid-19-living-guideline). The worldwide spread of SARS-CoV-2 virus revealed specific susceptibilities to the virus among the elderly and individuals with age-related syndromes. These populations were more likely to experience a hyperimmune response characterized by a treatment-resistant acute lung pathology accompanied by multiple organ failure. These observations underscore the interplay between the virus, the biology of aging, and outcomes observed in the most severe cases of SARS-CoV-2 infection. The ectoenzyme CD38 has been implicated in the process of "inflammaging" in aged tissues. In a current publication, Horenstein et al. present evidence to support the hypothesis that CD38 plays a central role in altered immunometabolism resulting from COVID-19 infection. The authors discuss a critical but underappreciated trifecta of CD38-mediated NAD+ metabolism, aging, and COVID-19 immune response and speculate that the CD38/NAD+ axis is a promising therapeutic target for this disease.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , COVID-19/physiopathology , Membrane Glycoproteins/metabolism , SARS-CoV-2 , ADP-ribosyl Cyclase 1/genetics , Aging , Gene Expression Regulation, Enzymologic , Humans , Membrane Glycoproteins/genetics , NAD/metabolism
9.
Front Immunol ; 12: 693269, 2021.
Article in English | MEDLINE | ID: covidwho-1389185

ABSTRACT

Chronic immune activation has been considered as the driving force for CD4+ T cell depletion in people infected with HIV-1. Interestingly, the normal immune profile of adult HIV-negative individuals living in Africa also exhibit chronic immune activation, reminiscent of that observed in HIV-1 infected individuals. It is characterized by increased levels of soluble immune activation markers, such as the cytokines interleukin (IL)-4, IL-10, TNF-α, and cellular activation markers including HLA-DR, CD-38, CCR5, coupled with reduced naïve and increased memory cells in CD4+ and CD8+ subsets. In addition, it is accompanied by low CD4+ T cell counts when compared to Europeans. There is also evidence that mononuclear cells from African infants secrete less innate cytokines than South and North Americans and Europeans in vitro. Chronic immune activation in Africans is linked to environmental factors such as parasitic infections and could be responsible for previously observed immune hypo-responsiveness to infections and vaccines. It is unclear whether the immunogenicity and effectiveness of anti-SARS-CoV-2 vaccines will also be reduced by similar mechanisms. A review of studies investigating this phenomenon is urgently required as they should inform the design and delivery for vaccines to be used in African populations.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , COVID-19 Vaccines/immunology , Immunogenicity, Vaccine/immunology , Lymphocyte Activation/immunology , SARS-CoV-2/immunology , ADP-ribosyl Cyclase 1/blood , Africa , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , COVID-19/prevention & control , HLA-DR Antigens/blood , Humans , Interleukin-10/blood , Interleukin-4/blood , Leukocytes, Mononuclear/metabolism , Membrane Glycoproteins/blood , Receptors, CCR5/blood , Tumor Necrosis Factor-alpha/blood
10.
Int Rev Cell Mol Biol ; 363: 203-269, 2021.
Article in English | MEDLINE | ID: covidwho-1212320

ABSTRACT

An increase in intracellular Ca2+ concentration ([Ca2+]i) regulates a plethora of functions in the cardiovascular (CV) system, including contraction in cardiomyocytes and vascular smooth muscle cells (VSMCs), and angiogenesis in vascular endothelial cells and endothelial colony forming cells. The sarco/endoplasmic reticulum (SR/ER) represents the largest endogenous Ca2+ store, which releases Ca2+ through ryanodine receptors (RyRs) and/or inositol-1,4,5-trisphosphate receptors (InsP3Rs) upon extracellular stimulation. The acidic vesicles of the endolysosomal (EL) compartment represent an additional endogenous Ca2+ store, which is targeted by several second messengers, including nicotinic acid adenine dinucleotide phosphate (NAADP) and phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2], and may release intraluminal Ca2+ through multiple Ca2+ permeable channels, including two-pore channels 1 and 2 (TPC1-2) and Transient Receptor Potential Mucolipin 1 (TRPML1). Herein, we discuss the emerging, pathophysiological role of EL Ca2+ signaling in the CV system. We describe the role of cardiac TPCs in ß-adrenoceptor stimulation, arrhythmia, hypertrophy, and ischemia-reperfusion injury. We then illustrate the role of EL Ca2+ signaling in VSMCs, where TPCs promote vasoconstriction and contribute to pulmonary artery hypertension and atherosclerosis, whereas TRPML1 sustains vasodilation and is also involved in atherosclerosis. Subsequently, we describe the mechanisms whereby endothelial TPCs promote vasodilation, contribute to neurovascular coupling in the brain and stimulate angiogenesis and vasculogenesis. Finally, we discuss about the possibility to target TPCs, which are likely to mediate CV cell infection by the Severe Acute Respiratory Disease-Coronavirus-2, with Food and Drug Administration-approved drugs to alleviate the detrimental effects of Coronavirus Disease-19 on the CV system.


Subject(s)
COVID-19 Drug Treatment , COVID-19/complications , Calcium Signaling/physiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Lysosomes/metabolism , SARS-CoV-2 , ADP-ribosyl Cyclase 1/metabolism , Animals , Brain/blood supply , Brain/metabolism , COVID-19/metabolism , Calcium Channels/metabolism , Cardiovascular Diseases/drug therapy , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Humans , Models, Cardiovascular , Myocytes, Cardiac/metabolism , NADP/analogs & derivatives , NADP/metabolism , Receptors, Adrenergic, beta/metabolism , Sarcoplasmic Reticulum/metabolism , Transient Receptor Potential Channels/metabolism
11.
Int Immunopharmacol ; 97: 107685, 2021 Aug.
Article in English | MEDLINE | ID: covidwho-1188659

ABSTRACT

BACKGROUND: The 2019 Coronavirus (COVID-19) pandemic poses a huge threat internationally; however, the role of the host immune system in the pathogenesis of COVID-19 is not well understood. METHODS: Cytokine and chemokine levels and characterisation of immune cell subsets from 20 COVID-19 cases after hospital admission (17 critically ill and 3 severe patients) and 16 convalescent patients were determined using a multiplex immunoassay and flow cytometry, respectively. RESULTS: IP-10, MCP-1, MIG, IL-6, and IL-10 levels were significantly higher in acute severe/critically ill patients with COVID-19, whereas were normal in patients who had reached convalescence. CD8 T cells in severe and critically ill COVID-19 patients expressed high levels of cytotoxic granules (granzyme B and perforin)and was hyperactivated as evidenced by the high proportions of CD38. Furthermore, the cytotoxic potential of natural killer (NK) cells, and the frequencies of myeloid dendritic cells and plasmacytoid dendritic cells was reduced in patients with severe and critical COVID-19; however, these dysregulations were found to be restored in convalescent phases. CONCLUSION: Thus, elicitation of the hyperactive cytokine-mediated inflammatory response, dysregulation of CD8 T and NK cells, and deficiency of host myeloid and plasmacytoid DCs, may contribute to COVID-19 pathogenesis and provide insights into potential therapeutic targets and strategies.


Subject(s)
COVID-19/blood , COVID-19/immunology , Convalescence , Inflammation/etiology , ADP-ribosyl Cyclase 1/blood , Acute Disease , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL2/blood , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Critical Illness , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/immunology , Dendritic Cells/immunology , Female , Granzymes/metabolism , Humans , Interleukin-10/blood , Interleukin-6/blood , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , Male , Membrane Glycoproteins/blood , Middle Aged , Perforin/metabolism
12.
J Immunol ; 206(7): 1478-1482, 2021 04 01.
Article in English | MEDLINE | ID: covidwho-1073559

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has become pandemic. Cytokine release syndrome occurring in a minority of SARS-CoV-2 infections is associated with severe disease and high mortality. We profiled the composition, activation, and proliferation of T cells in 20 patients with severe or critical COVID-19 and 40 matched healthy controls by flow cytometry. Unsupervised hierarchical cluster analysis based on 18 T cell subsets resulted in separation of healthy controls and COVID-19 patients. Compared to healthy controls, patients suffering from severe and critical COVID-19 had increased frequencies of activated and proliferating CD38+Ki67+ CD4+ and CD8+ T cells, suggesting active antiviral T cell defense. Frequencies of CD38+Ki67+ Th1 and CD4+ cells correlated negatively with plasma IL-6. Thus, our data suggest that patients suffering from COVID-19 have a distinct T cell composition that is potentially modulated by IL-6.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Immunity, Cellular , SARS-CoV-2/immunology , Th1 Cells/immunology , ADP-ribosyl Cyclase 1/immunology , Adult , CD8-Positive T-Lymphocytes/pathology , COVID-19/epidemiology , COVID-19/pathology , Female , Humans , Immunophenotyping , Interleukin-6/immunology , Ki-67 Antigen/immunology , Male , Membrane Glycoproteins/immunology , Pandemics , Retrospective Studies , Th1 Cells/pathology
13.
Hum Immunol ; 82(3): 170-176, 2021 Mar.
Article in English | MEDLINE | ID: covidwho-1065106

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19) is affecting the whole world and threatening human health. We aim to investigate the immunological characteristics of monocytes in critical patients with COVID-19. METHODS: The number and immune status of monocytes were detected by flow cytometry in 32 COVID-19 patients and 18 healthy individuals. RESULTS: In critical patients with COVID-19, the absolute number of total monocytes and CD16- monocytes was significantly decreased but CD16+ pro-inflammatory monocytes was increased compared to healthy controls. Antigen presentation potential of monocytes, as measured by HLA-DR expression, was suppressed, while their inflammatory phenotype (CD38 expression) was enhanced. Cytokine levels showed sustained increases in critical patients. And the levels of IL-6 were positively correlated with CD16+ monocytes number. IL-6 and IL-10 levels were negatively correlated with HLA-DR expression of monocytes. During the recovery of COVID-19 patients, the count and immune status of monocyte subsets were restored by degrees. HLA-DR+ monocytes possessed good sensitivity and specificity for predicting the incidence of critical patients with COVID-19. CONCLUSIONS: In critical patients with COVID-19, decline in number and HLA-DR expression of monocytes might lead to decreased antigen presentation potential and thus immunosuppression, while increased CD16+ pro-inflammatory monocytes might mediate hyperinflammation. HLA-DR+ monocytes might be a meaningful assisted indicator to predict the incidence of critical patients with COVID-19.


Subject(s)
COVID-19/immunology , Monocytes/immunology , ADP-ribosyl Cyclase 1/immunology , Aged , Antigen Presentation , COVID-19/blood , Case-Control Studies , Cytokines/immunology , Female , Flow Cytometry , HLA-DR Antigens/immunology , Humans , Inflammation/immunology , Male , Middle Aged , Phenotype , Receptors, IgG/immunology
14.
Front Immunol ; 11: 596553, 2020.
Article in English | MEDLINE | ID: covidwho-979020

ABSTRACT

The severity of SARS-CoV-2 infection has been related to uncontrolled inflammatory innate responses and impaired adaptive immune responses mostly due to exhausted T lymphocytes and lymphopenia. In this work we have characterized the nature of the lymphopenia and demonstrate a set of factors that hinder the effective control of virus infection and the activation and arming of effector cytotoxic T CD8 cells and showing signatures defining a high-risk population. We performed immune profiling of the T helper (Th) CD4+ and T CD8+ cell compartments in peripheral blood of 144 COVID-19 patients using multiparametric flow cytometry analysis. On the one hand, there was a consistent lymphopenia with an overrepresentation of non-functional T cells, with an increased percentage of naive Th cells (CD45RA+, CXCR3-, CCR4-, CCR6-, CCR10-) and persistently low frequency of markers associated with Th1, Th17, and Th1/Th17 memory-effector T cells compared to healthy donors. On the other hand, the most profound alteration affected the Th1 subset, which may explain the poor T cells responses and the persistent blood virus load. Finally, the decrease in Th1 cells may also explain the low frequency of CD4+ and CD8+ T cells that express the HLA-DR and CD38 activation markers observed in numerous patients who showed minimal or no lymphocyte activation response. We also identified the percentage of HLA-DR+CD4+ T cells, PD-1+CD+4/CD8+ T cells in blood, and the neutrophil/lymphocyte ratio as useful factors for predicting critical illness and fatal outcome in patients with confirmed COVID-19.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , SARS-CoV-2/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , ADP-ribosyl Cyclase 1/immunology , ADP-ribosyl Cyclase 1/metabolism , Aged , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , COVID-19/virology , Cell Differentiation/immunology , Female , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Humans , Lymphocyte Activation/immunology , Male , Middle Aged , Prospective Studies , SARS-CoV-2/physiology , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
15.
Med Hypotheses ; 144: 110044, 2020 Nov.
Article in English | MEDLINE | ID: covidwho-623607

ABSTRACT

The SARS-CoV-2 hyperinflammatory response is associated with high mortality. This hypothesis suggests that a deficiency of nicotinamide adenine dinucleotide (NAD+) may be the primary factor related to the SARS-Cov-2 disease spectrum and the risk for mortality, as subclinical nutritional deficiencies may be unmasked by any significant increase in oxidative stress. NAD+ levels decline with age and are also reduced in conditions associated with oxidative stress as occurs with hypertension, diabetes and obesity. These groups have also been observed to have high mortality following infection with COVID-19. Further consumption of NAD+ in a pre-existent depleted state is more likely to cause progression to the hyperinflammatory stage of the disease through its limiting effects on the production of SIRT1. This provides a unifying hypothesis as to why these groups are at high risk of mortality and suggests that nutritional support with NAD+ and SIRT1 activators, could minimise disease severity if administered prophylactically and or therapeutically. The significance of this, if proven, has far-reaching consequences in the management of COVID-19 especially in third world countries, where resources and finances are limited.


Subject(s)
COVID-19/immunology , Diabetes Mellitus, Type 2/complications , NAD/deficiency , Obesity/complications , Sirtuin 1/immunology , ADAM17 Protein/immunology , ADP-ribosyl Cyclase 1/immunology , Age Factors , Aged , Aging , COVID-19/mortality , Diabetes Mellitus, Type 2/immunology , Disease Progression , Disease Susceptibility , Humans , Inflammation , Membrane Glycoproteins/immunology , NAD/chemistry , Obesity/immunology , Oxidative Stress , Protein Binding , Virus Replication , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL