Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Comput Struct Biotechnol J ; 19: 976-988, 2021.
Article in English | MEDLINE | ID: covidwho-2266096

ABSTRACT

Chemokines are crucial inflammatory mediators needed during an immune response to clear pathogens. However, their excessive release is the main cause of hyperinflammation. In the recent COVID-19 outbreak, chemokines may be the direct cause of acute respiratory disease syndrome, a major complication leading to death in about 40% of severe cases. Several clinical investigations revealed that chemokines are directly involved in the different stages of SARS-CoV-2 infection. Here, we review the role of chemokines and their receptors in COVID-19 pathogenesis to better understand the disease immunopathology which may aid in developing possible therapeutic targets for the infection.

2.
Scandinavian Journal of Immunology ; 97(1), 2023.
Article in English | ProQuest Central | ID: covidwho-2193234

ABSTRACT

COVID‐19, which emerged in December 2019 and continues to wreak havoc, has led to the death of many people around the world. In this study, we aimed to uncover the variables underlying the exacerbation of the disease by considering the changes in T cell subsets in adults and juveniles with different disease severity of COVID‐19. Peripheral blood samples of 193 patients (128 adults and 65 juveniles) diagnosed with COVID‐19 were evaluated in a flow cytometer, and a broad T cell profile was revealed by examining T cell subsets in terms of exhaustion and senescence. We found remarkable differences in the effector memory (EM;CD45RA−CCR7−) cell subsets of severe pneumonia cases. The frequencies of EM2 CD4+ T, EM3 CD4+ T, EM3 CD8+ T, EM2 DN T and EM3 DN T cells were found to increase in severe pneumonia cases. Consistently, these cells were found in juveniles and uncomplicated adults in similar or lower proportions to healthy controls. The findings of our study provide a view of the T cell profile that may underlie differences in the course of COVID‐19 cases in juveniles and adults and may provide new insights into the development of effective treatment strategies.

3.
Front Immunol ; 13: 931388, 2022.
Article in English | MEDLINE | ID: covidwho-2141951

ABSTRACT

Intracranial inoculation of the neuroadapted JHM strain of mouse hepatitis virus (JHMV) into susceptible strains of mice results in acute encephalomyelitis followed by a cimmune-mediated demyelination similar to the human demyelinating disease multiple sclerosis (MS). JHMV infection of transgenic mice in which expression of the neutrophil chemoattractant chemokine CXCL1 is under the control of a tetracycline-inducible promoter active within GFAP-positive cells results in sustained neutrophil infiltration in the central nervous system (CNS) that correlates with an increase in spinal cord demyelination. We used single cell RNA sequencing (scRNAseq) and flow cytometry to characterize molecular and cellular changes within the CNS associated with increased demyelination in transgenic mice compared to control animals. These approaches revealed the presence of activated neutrophils as determined by expression of mRNA transcripts associated with neutrophil effector functions, including CD63, MMP9, S100a8, S100a9, and ASPRV1, as well as altered neutrophil morphology and protein expression. Collectively, these findings reveal insight into changes in the profile of neutrophils associated with increased white matter damage in mice persistently infected with a neurotropic coronavirus.


Subject(s)
Demyelinating Diseases , Multiple Sclerosis , Murine hepatitis virus , White Matter , Animals , Central Nervous System , Chemokine CXCL1/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/metabolism , Neutrophils/metabolism , RNA, Messenger , Tetracyclines , White Matter/metabolism
4.
Viruses ; 14(9)2022 08 28.
Article in English | MEDLINE | ID: covidwho-2006222

ABSTRACT

BACKGROUND: The adaptive antiviral immune response requires interaction between CD8+ T cells, dendritic cells, and Th1 cells for controlling SARS-CoV-2 infection, but the data regarding the role of CD8+ T cells in the acute phase of COVID-19 and post-COVID-19 syndrome are still limited. METHODS: . Peripheral blood samples collected from patients with acute COVID-19 (n = 71), convalescent subjects bearing serum SARS-CoV-2 N-protein-specific IgG antibodies (n = 51), and healthy volunteers with no detectable antibodies to any SARS-CoV-2 proteins (HC, n = 46) were analyzed using 10-color flow cytometry. RESULTS: Patients with acute COVID-19 vs. HC and COVID-19 convalescents showed decreased absolute numbers of CD8+ T cells, whereas the frequency of CM and TEMRA CD8+ T cells in acute COVID-19 vs. HC was elevated. COVID-19 convalescents vs. HC had increased naïve and CM cells, whereas TEMRA cells were decreased compared to HC. Cell-surface CD57 was highly expressed by the majority of CD8+ T cells subsets during acute COVID-19, but convalescents had increased CD57 on 'naïve', CM, EM4, and pE1 2-3 months post-symptom onset. CXCR5 expression was altered in acute and convalescent COVID-19 subjects, whereas the frequencies of CXCR3+ and CCR4+ cells were decreased in both patient groups vs. HC. COVID-19 convalescents had increased CCR6-expressing CD8+ T cells. Moreover, CXCR3+CCR6- Tc1 cells were decreased in patients with acute COVID-19 and COVID-19 convalescents, whereas Tc2 and Tc17 levels were increased compared to HC. Finally, IL-27 negatively correlated with the CCR6+ cells in acute COVID-19 patients. CONCLUSIONS: We described an abnormal CD8+ T cell profile in COVID-19 convalescents, which resulted in lower frequencies of effector subsets (TEMRA and Tc1), higher senescent state (upregulated CD57 on 'naïve' and memory cells), and higher frequencies of CD8+ T cell subsets expressing lung tissue and mucosal tissue homing molecules (Tc2, Tc17, and Tc17.1). Thus, our data indicate that COVID-19 can impact the long-term CD8+ T cell immune response.


Subject(s)
COVID-19 , Interleukin-27 , Antiviral Agents/metabolism , CD8-Positive T-Lymphocytes , COVID-19/complications , Humans , Immunoglobulin G , SARS-CoV-2 , Post-Acute COVID-19 Syndrome
5.
Front Immunol ; 13: 834862, 2022.
Article in English | MEDLINE | ID: covidwho-1775666

ABSTRACT

Respiratory viral infections with SARS-CoV-2 and influenza viruses commonly induce a strong infiltration of immune cells into the human lung, with potential detrimental effects on the integrity of the lung tissue. Despite comprising the largest fractions of circulating lymphocytes in the lung, rather little is known about how peripheral blood natural killer (NK) cell and T cell subsets are equipped for lung-homing in COVID-19 and influenza. Here, we provide a detailed comparative analysis of NK cells and T cells in patients infected with SARS-CoV-2 or influenza virus, focusing on the protein and gene expression of chemokine receptors known to be involved in recruitment to the lung. For this, we used 28-colour flow cytometry as well as re-analysis of a publicly available single-cell RNA-seq dataset from bronchoalveolar lavage (BAL) fluid. Frequencies of NK cells and T cells expressing CXCR3, CXCR6, and CCR5 were altered in peripheral blood of COVID-19 and influenza patients, in line with increased transcript expression of CXCR3, CXCR6, and CCR5 and their respective ligands in BAL fluid. NK cells and T cells expressing lung-homing receptors displayed stronger phenotypic signs of activation compared to cells lacking lung-homing receptors, and activation was overall stronger in influenza compared to COVID-19. Together, our results indicate a role for CXCR3+, CXCR6+, and/or CCR5+ NK cells and T cells that potentially migrate to the lungs in moderate COVID-19 and influenza patients, identifying common targets for future therapeutic interventions in respiratory viral infections.


Subject(s)
COVID-19 , Influenza, Human , Gene Expression , Humans , Influenza, Human/metabolism , Killer Cells, Natural , Lung , SARS-CoV-2 , T-Lymphocyte Subsets
6.
Cancer Metastasis Rev ; 41(1): 147-172, 2022 03.
Article in English | MEDLINE | ID: covidwho-1635411

ABSTRACT

We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.


Subject(s)
COVID-19 , Neoplasms , Animals , Biology , Blood Platelets/metabolism , Hemostasis , Humans , Mammals , Neoplasms/metabolism , SARS-CoV-2
7.
Expert Rev Clin Immunol ; 18(1): 57-66, 2022 01.
Article in English | MEDLINE | ID: covidwho-1577592

ABSTRACT

INTRODUCTION: Coronaviruses are a large family of positive-stranded nonsegmented RNA viruses with genomes of 26-32 kilobases in length. Human coronaviruses are commonly associated with mild respiratory illness; however, the past three decades have seen the emergence of severe acute respiratory coronavirus (SARS-CoV), middle eastern respiratory coronavirus (MERS-CoV), and SARS-CoV-2 which is the etiologic agent for COVID-19. Severe forms of COVID-19 include acute respiratory distress syndrome (ARDS) associated with cytokine release syndrome that can culminate in multiorgan failure and death. Among the proinflammatory factors associated with severe COVID-19 are the chemokines CCL2, CCL3, CXCL8, and CXCL10. Infection of susceptible mice with murine coronaviruses, such as mouse hepatitis virus (MHV), elicits a similar chemokine response profile as observed in COVID-19 patients and these in vivo models have been informative and show that targeting chemokines reduces the severity of inflammation in target organs. AREAS COVERED: PubMed was used using keywords: Chemokines and coronaviruses; Chemokines and mouse hepatitis virus; Chemokines and COVID-19. Clinicaltrials.gov was used using keywords: COVID-19 and chemokines; COVID-19 and cytokines; COVID-19 and neutrophil. EXPERT OPINION: Chemokines and chemokine receptors are clinically relevant therapeutic targets for reducing coronavirus-induced inflammation.


Subject(s)
COVID-19 , Receptors, Chemokine , Animals , Chemokines , Cytokine Release Syndrome , Humans , Mice , SARS-CoV-2
8.
Immunity ; 54(2): 340-354.e6, 2021 02 09.
Article in English | MEDLINE | ID: covidwho-1071479

ABSTRACT

Cellular and humoral immunity to SARS-CoV-2 is critical to control primary infection and correlates with severity of disease. The role of SARS-CoV-2-specific T cell immunity, its relationship to antibodies, and pre-existing immunity against endemic coronaviruses (huCoV), which has been hypothesized to be protective, were investigated in 82 healthy donors (HDs), 204 recovered (RCs), and 92 active COVID-19 patients (ACs). ACs had high amounts of anti-SARS-CoV-2 nucleocapsid and spike IgG but lymphopenia and overall reduced antiviral T cell responses due to the inflammatory milieu, expression of inhibitory molecules (PD-1, Tim-3) as well as effector caspase-3, -7, and -8 activity in T cells. SARS-CoV-2-specific T cell immunity conferred by polyfunctional, mainly interferon-γ-secreting CD4+ T cells remained stable throughout convalescence, whereas humoral responses declined. Immune responses toward huCoV in RCs with mild disease and strong cellular SARS-CoV-2 T cell reactivity imply a protective role of pre-existing immunity against huCoV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , COVID-19/immunology , Immunity, Cellular/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Humans , Immunity, Humoral/immunology , Male , Middle Aged , SARS-CoV-2/immunology , Young Adult
9.
Viruses ; 13(2)2021 01 28.
Article in English | MEDLINE | ID: covidwho-1060540

ABSTRACT

There have been reports of neurological abnormalities associated with the Zika virus (ZIKV), such as congenital Zika syndrome (CZS) in children born to mothers infected during pregnancy. We investigated how the immune response to ZIKV during pregnancy is primed and conduct a thorough evaluation of the inflammatory and cytotoxic profiles as well as the expression of CCR5 and CX3CR1. We compared the reactivity of T cells to ZIKV peptides in convalescent mothers infected during pregnancy. The child's clinical outcome (i.e., born with or without CZS) was taken to be the variable. The cells were stimulated in vitro with ZIKV peptides and evaluated using the ELISPOT and flow cytometry assays. After in vitro stimulation with ZIKV peptides, we observed a tendency toward a higher Interferon gamma (IFN-γ)-producing T cell responses in mothers who had asymptomatic children and a higher CD107a expression in T cells in mothers who had children with CZS. We found a higher frequency of T cells expressing CD107a+ and co-expressing CX3CR1+CCR5+, which is much clearer in the T cells of mothers who had CZS children. We suggest that this differential profile influenced the clinical outcome of babies. These data need to be further investigated, including the evaluation of other ZIKV peptides and markers and functional assays.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Pregnancy Complications, Infectious/immunology , Receptors, CCR5/metabolism , T-Lymphocytes/immunology , Zika Virus Infection/immunology , Adult , Cross-Sectional Studies , Cytotoxicity, Immunologic , Female , Humans , Infant , Interferon-gamma/metabolism , Lysosome-Associated Membrane Glycoproteins/metabolism , Pregnancy , Pregnancy Outcome , T-Lymphocytes/metabolism , Young Adult , Zika Virus/immunology
10.
Eur J Pharm Sci ; 155: 105537, 2020 Dec 01.
Article in English | MEDLINE | ID: covidwho-739808

ABSTRACT

Chemokine receptors are key regulators of cell migration in terms of immunity and inflammation. Among these, CCR5 and CXCR4 play pivotal roles in cancer metastasis and HIV-1 transmission and infection. They act as essential co-receptors for HIV and furnish a route to the cell entry. In particular, inhibition of either CCR5 or CXCR4 leads very often the virus to shift to a more virulent dual-tropic strain. Therefore, dual receptor inhibition might improve the therapeutic strategies against HIV. In this study, we aimed to discover selective CCR5, CXCR4, and dual CCR5/CXCR4 antagonists using both receptor- and ligand-based computational methods. We employed this approach to fully incorporate the interaction attributes of the binding pocket together with molecular dynamics (MD) simulations and binding free energy calculations. The best hits were evaluated for their anti-HIV-1 activity against CXCR4- and CCR5-specific NL4.3 and BaL strains. Moreover, the Ca2+ mobilization assay was used to evaluate their antagonistic activity. From the 27 tested compounds, three were identified as inhibitors: compounds 27 (CCR5), 6 (CXCR4) and 3 (dual) with IC50 values ranging from 10.64 to 64.56 µM. The binding mode analysis suggests that the active compounds form a salt bridge with the glutamates and π-stacking interactions with the aromatic side chains binding site residues of the respective co-receptor. The presented hierarchical virtual screening approach provides essential aspects in identifying potential antagonists in terms of selectivity against a specific co-receptor. The compounds having multiple heterocyclic nitrogen atoms proved to be relatively more specific towards CXCR4 inhibition as compared to CCR5. The identified compounds serve as a starting point for further development of HIV entry inhibitors through synthesis and quantitative structure-activity relationship studies.


Subject(s)
HIV Fusion Inhibitors , HIV Infections , HIV-1 , Binding Sites , CCR5 Receptor Antagonists/pharmacology , HIV Infections/drug therapy , Humans , Ligands , Receptors, CCR5
SELECTION OF CITATIONS
SEARCH DETAIL