Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article in English | MEDLINE | ID: covidwho-1667193

ABSTRACT

Elastin represents the structural component of the extracellular matrix providing elastic recoil to tissues such as skin, blood vessels and lungs. Elastogenic cells secrete soluble tropoelastin monomers into the extracellular space where these monomers associate with other matrix proteins (e.g., microfibrils and glycoproteins) and are crosslinked by lysyl oxidase to form insoluble fibres. Once elastic fibres are formed, they are very stable, highly resistant to degradation and have an almost negligible turnover. However, there are circumstances, mainly related to inflammatory conditions, where increased proteolytic degradation of elastic fibres may lead to consequences of major clinical relevance. In severely affected COVID-19 patients, for instance, the massive recruitment and activation of neutrophils is responsible for the profuse release of elastases and other proteolytic enzymes which cause the irreversible degradation of elastic fibres. Within the lungs, destruction of the elastic network may lead to the permanent impairment of pulmonary function, thus suggesting that elastases can be a promising target to preserve the elastic component in COVID-19 patients. Moreover, intrinsic and extrinsic factors additionally contributing to damaging the elastic component and to increasing the spread and severity of SARS-CoV-2 infection are reviewed.


Subject(s)
COVID-19/metabolism , Elastin/physiology , Extracellular Matrix/physiology , Animals , Elastic Tissue/metabolism , Elastin/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Traps/metabolism , Fibrillins/metabolism , Humans , Lung/pathology , Microfibrils/metabolism , Microfilament Proteins/metabolism , Neutrophils , Protein-Lysine 6-Oxidase/metabolism , SARS-CoV-2/pathogenicity , Tropoelastin/metabolism
2.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: covidwho-1383578

ABSTRACT

Proline-glycine-proline (PGP) and its acetylated form (Ac-PGP) are neutrophil chemoattractants generated by collagen degradation, and they have been shown to play a role in chronic inflammatory disease. However, the mechanism for matrikine regulation in acute inflammation has not been well established. Here, we show that these peptides are actively transported from the lung by the oligopeptide transporter, PEPT2. Following intratracheal instillation of Ac-PGP in a mouse model, there was a rapid decline in concentration of the labeled peptide in the bronchoalveolar lavage (BAL) over time and redistribution to extrapulmonary sites. In vitro knockdown of the PEPT2 transporter in airway epithelia or use of a competitive inhibitor of PEPT2, cefadroxil, significantly reduced uptake of Ac-PGP. Animals that received intratracheal Ac-PGP plus cefadroxil had higher levels of Ac-PGP in BAL and lung tissue. Utilizing an acute LPS-induced lung injury model, we demonstrate that PEPT2 blockade enhanced pulmonary Ac-PGP levels and lung inflammation. We further validated this effect using clinical samples from patients with acute lung injury in coculture with airway epithelia. This is the first study to our knowledge to determine the in vitro and in vivo significance of active matrikine transport as a mechanism of modulating acute inflammation and to demonstrate that it may serve as a potential therapeutic target.


Subject(s)
Acute Lung Injury/immunology , COVID-19 , Cefadroxil/pharmacology , Inflammation/metabolism , Oligopeptides , Proline/analogs & derivatives , Symporters , Animals , Anti-Bacterial Agents/pharmacology , Biological Transport, Active/immunology , COVID-19/immunology , COVID-19/metabolism , Cells, Cultured , Chemotactic Factors/immunology , Chemotactic Factors/pharmacology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Extracellular Matrix , Extracellular Matrix Proteins/metabolism , Humans , Mice , Oligopeptides/immunology , Oligopeptides/pharmacology , Proline/immunology , Proline/pharmacology , Symporters/antagonists & inhibitors , Symporters/metabolism
3.
Nutrients ; 13(6)2021 Jun 09.
Article in English | MEDLINE | ID: covidwho-1264499

ABSTRACT

It has recently been hypothesized that vitamin K could play a role in COVID-19. We aimed to test the hypotheses that low vitamin K status is a common characteristic of patients hospitalized with COVID-19 compared to population controls and that low vitamin K status predicts mortality in COVID-19 patients. In a cohort of 138 COVID-19 patients and 138 population controls, we measured plasma dephosphorylated-uncarboxylated Matrix Gla Protein (dp-ucMGP), which reflects the functional vitamin K status in peripheral tissue. Forty-three patients died within 90 days from admission. In patients, levels of dp-ucMGP differed significantly between survivors (mean 877; 95% CI: 778; 995) and non-survivors (mean 1445; 95% CI: 1148; 1820). Furthermore, levels of dp-ucMGP (pmol/L) were considerably higher in patients (mean 1022; 95% CI: 912; 1151) compared to controls (mean 509; 95% CI: 485; 540). Cox regression survival analysis showed that increasing levels of dp-ucMGP (reflecting low vitamin K status) were associated with higher mortality risk (sex- and age-adjusted hazard ratio per doubling of dp-ucMGP was 1.49, 95% CI: 1.03; 2.24). The association attenuated and became statistically insignificant after adjustment for co-morbidities (sex, age, CVD, diabetes, BMI, and eGFR adjusted hazard ratio per doubling of dp-ucMGP was 1.22, 95% CI: 0.82; 1.80). In conclusion, we found that low vitamin K status was associated with mortality in patients with COVID-19 in sex- and age-adjusted analyses, but not in analyses additionally adjusted for co-morbidities. Randomized clinical trials would be needed to clarify a potential role, if any, of vitamin K in the course of COVID-19.


Subject(s)
COVID-19/mortality , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Hospitalization , Vitamin K Deficiency/mortality , Vitamin K/blood , Adult , Aged , Biomarkers/blood , Blood Coagulation , COVID-19/complications , COVID-19/metabolism , Calcium-Binding Proteins/blood , Cohort Studies , Extracellular Matrix Proteins/blood , Female , Hospital Mortality , Humans , Male , Middle Aged , Proportional Hazards Models , Regression Analysis , SARS-CoV-2 , Thrombosis/metabolism , Vitamin K Deficiency/blood , Vitamin K Deficiency/complications , Young Adult
4.
Br J Nutr ; 126(2): 191-198, 2021 07 28.
Article in English | MEDLINE | ID: covidwho-1261982

ABSTRACT

Coronavirus disease 2019 (Covid-19), caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2, exerts far-reaching effects on public health and socio-economic welfare. The majority of infected individuals have mild to moderate symptoms, but a significant proportion develops respiratory failure due to pneumonia. Thrombosis is another frequent manifestation of Covid-19 that contributes to poor outcomes. Vitamin K plays a crucial role in the activation of both pro- and anticlotting factors in the liver and the activation of extrahepatically synthesised protein S which seems to be important in local thrombosis prevention. However, the role of vitamin K extends beyond coagulation. Matrix Gla protein (MGP) is a vitamin K-dependent inhibitor of soft tissue calcification and elastic fibre degradation. Severe extrahepatic vitamin K insufficiency was recently demonstrated in Covid-19 patients, with high inactive MGP levels correlating with elastic fibre degradation rates. This suggests that insufficient vitamin K-dependent MGP activation leaves elastic fibres unprotected against SARS-CoV-2-induced proteolysis. In contrast to MGP, Covid-19 patients have normal levels of activated factor II, in line with previous observations that vitamin K is preferentially transported to the liver for activation of procoagulant factors. We therefore expect that vitamin K-dependent endothelial protein S activation is also compromised, which would be compatible with enhanced thrombogenicity. Taking these data together, we propose a mechanism of pneumonia-induced vitamin K depletion, leading to a decrease in activated MGP and protein S, aggravating pulmonary damage and coagulopathy, respectively. Intervention trials should be conducted to assess whether vitamin K administration plays a role in the prevention and treatment of severe Covid-19.


Subject(s)
COVID-19/pathology , Lung/physiopathology , SARS-CoV-2 , Thromboembolism/prevention & control , Thrombosis/prevention & control , Vitamin K Deficiency/metabolism , Vitamin K/metabolism , COVID-19/complications , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Protein S/metabolism , Thromboembolism/etiology , Thrombosis/etiology , Vitamin K/antagonists & inhibitors , Vitamin K Deficiency/etiology
5.
Mol Biol Cell ; 32(14): 1273-1282, 2021 07 01.
Article in English | MEDLINE | ID: covidwho-1233836

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that has triggered global health and economic crises. Here we report the effects of SARS-CoV-2 infection on peroxisomes of human cell lines Huh-7 and SK-N-SH. Peroxisomes undergo dramatic changes in morphology in SARS-CoV-2-infected cells. Rearrangement of peroxisomal membranes is followed by redistribution of peroxisomal matrix proteins to the cytosol, resulting in a dramatic decrease in the number of mature peroxisomes. The SARS-CoV-2 ORF14 protein was shown to interact physically with human PEX14, a peroxisomal membrane protein required for matrix protein import and peroxisome biogenesis. Given the important roles of peroxisomes in innate immunity, SARS-CoV-2 may directly target peroxisomes, resulting in loss of peroxisome structural integrity, matrix protein content and ability to function in antiviral signaling.


Subject(s)
Peroxisomes/virology , Animals , Cell Line , Cell Membrane/pathology , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Membrane Proteins/metabolism , Peroxisomes/metabolism , Peroxisomes/pathology , Phosphoproteins/metabolism , Repressor Proteins/metabolism , SARS-CoV-2/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL