Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Front Immunol ; 13: 1021928, 2022.
Article in English | MEDLINE | ID: covidwho-2123417

ABSTRACT

ACE2 and TMPRSS2 are crucial for SARS-CoV-2 entry into the cell. Although ACE2 facilitates viral entry, its loss leads to promoting the devastating clinical symptoms of COVID-19 disease. Thus, enhanced ACE2/TMPRSS2 expression is likely to increase predisposition of target cells to SARS-CoV-2 infection. However, little evidence existed about the biological kinetics of these two enzymes and whether dexamethasone treatment modulates their expression. Here, we show that the expression of ACE2 at the protein and mRNA levels was significantly higher in the lung and heart tissues of neonatal compared to adult mice. However, the expression of TMPRSS2 was developmentally regulated. Our results may introduce a novel concept for the reduced susceptibility of the young to SARS-CoV-2 infection. Moreover, ACE2 expression but not TMPRSS2 was upregulated in adult female lungs compared to their male counterparts. Interestingly, the ACE2 and TMPRSS2 expressions were upregulated by dexamethasone treatment in the lung and heart tissues in both neonatal and adult mice. Furthermore, our findings provide a novel mechanism for the observed differential therapeutic effects of dexamethasone in COVID-19 patients. As such, dexamethasone exhibits different therapeutic effects depending on the disease stage. This was supported by increased ACE2/TMPRSS2 expression and subsequently enhanced infection of normal human bronchial epithelial cells (NHBE) and Vero E6 cells with SARS-CoV-2 once pre-treated with dexamethasone. Therefore, our results suggest that individuals who take dexamethasone for other clinical conditions may become more prone to SARS-CoV-2 infection.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 Drug Treatment , Humans , Male , Female , Mice , Animals , Angiotensin-Converting Enzyme 2/genetics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2 , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Serine Endopeptidases/genetics
2.
Stem Cell Reports ; 16(5): 1165-1181, 2021 05 11.
Article in English | MEDLINE | ID: covidwho-1225410

ABSTRACT

SARS-CoV-2 infection is associated with lower blood oxygen levels, even in patients without hypoxia requiring hospitalization. This discordance illustrates the need for a more unifying explanation as to whether SARS-CoV-2 directly or indirectly affects erythropoiesis. Here, we show significantly enriched CD71+ erythroid precursors/progenitors in the blood circulation of COVID-19 patients. We found that these cells have distinctive immunosuppressive properties. In agreement, we observed a strong negative correlation between the frequency of these cells with T and B cell proportions in COVID-19 patients. The expansion of these CD71+ erythroid precursors/progenitors was negatively correlated with the hemoglobin levels. A subpopulation of abundant erythroid cells, CD45+ CD71+ cells, co-express ACE2, TMPRSS2, CD147, and CD26, and these can be infected with SARS-CoV-2. In turn, pre-treatment of erythroid cells with dexamethasone significantly diminished ACE2/TMPRSS2 expression and subsequently reduced their infectivity with SARS-CoV-2. This provides a novel insight into the impact of SARS-CoV-2 on erythropoiesis and hypoxia seen in COVID-19 patients.


Subject(s)
Adaptive Immunity/immunology , COVID-19/pathology , Erythroid Precursor Cells/virology , Erythropoiesis/physiology , Hemoglobins/analysis , Oxygen/blood , Adolescent , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , COVID-19/immunology , Dexamethasone/pharmacology , Erythroid Precursor Cells/immunology , Female , Humans , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Middle Aged , SARS-CoV-2/immunology , Serine Endopeptidases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Young Adult
3.
Front Immunol ; 11: 597433, 2020.
Article in English | MEDLINE | ID: covidwho-983709

ABSTRACT

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Subject(s)
Antigens, CD/immunology , Erythroid Precursor Cells , Immunosuppression Therapy , Listeria monocytogenes/immunology , Listeriosis , Receptors, Transferrin/immunology , Animals , Animals, Newborn , COVID-19/immunology , COVID-19/pathology , Erythroid Precursor Cells/immunology , Erythroid Precursor Cells/pathology , Erythroid Precursor Cells/transplantation , Female , Heterografts , Humans , Infant, Newborn , Listeriosis/immunology , Listeriosis/pathology , Listeriosis/therapy , Male , Mice , Mice, Inbred BALB C , SARS-CoV-2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL