Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Glycobiology ; 33(6): 476-489, 2023 06 21.
Article in English | MEDLINE | ID: covidwho-2304778

ABSTRACT

The COVID-19 global pandemic has underscored the need to understand how viruses and other pathogens are able to infect and replicate within the respiratory system. Recent studies have highlighted the role of highly O-glycosylated mucins in the protection of the respiratory system as well as how mucin-type O-glycosylation may be able to modify viral infectivity. Therefore, we set out to identify the specific genes controlling mucin-type O-glycosylation throughout the mouse respiratory system as well as determine how their expression and the expression of respiratory mucins is influenced by infection or injury. Here, we show that certain mucins and members of the Galnt family are abundantly expressed in specific respiratory tissues/cells and demonstrate unique patterns of O-glycosylation across diverse respiratory tissues. Moreover, we find that the expression of certain Galnts and mucins is altered during lung infection and injury in experimental mice challenged with infectious agents, toxins, and allergens. Finally, we examine gene expression changes of Galnts and mucins in a mouse model of SARS-CoV-2 infection. Our work provides foundational knowledge regarding the specific expression of Galnt enzyme family members and mucins throughout the respiratory system, and how their expression is altered upon lung infection and injury.


Subject(s)
COVID-19 , Mucins , Animals , Mice , Mucins/genetics , Mucins/metabolism , Glycosylation , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Respiratory System/metabolism
2.
Adv Sci (Weinh) ; 9(32): e2203898, 2022 11.
Article in English | MEDLINE | ID: covidwho-2118630

ABSTRACT

Mucus is a self-healing gel that lubricates the moist epithelium and provides protection against viruses by binding to viruses smaller than the gel's mesh size and removing them from the mucosal surface by active mucus turnover. As the primary nonaqueous components of mucus (≈0.2%-5%, wt/v), mucins are critical to this function because the dense arrangement of mucin glycans allows multivalence of binding. Following nature's example, bovine submaxillary mucins (BSMs) are assembled into "mucus-like" gels (5%, wt/v) by dynamic covalent crosslinking reactions. The gels exhibit transient liquefaction under high shear strain and immediate self-healing behavior. This study shows that these material properties are essential to provide lubricity. The gels efficiently reduce human immunodeficiency virus type 1 (HIV-1) and genital herpes virus type 2 (HSV-2) infectivity for various types of cells. In contrast, simple mucin solutions, which lack the structural makeup, inhibit HIV-1 significantly less and do not inhibit HSV-2. Mechanistically, the prophylaxis of HIV-1 infection by BSM gels is found to be that the gels trap HIV-1 by binding to the envelope glycoprotein gp120 and suppress cytokine production during viral exposure. Therefore, the authors believe the gels are promising for further development as personal lubricants that can limit viral transmission.


Subject(s)
HIV-1 , Animals , Cattle , Humans , HIV-1/metabolism , Herpesvirus 2, Human/metabolism , Mucins/metabolism , Gels , Mucus/metabolism
3.
Folia Microbiol (Praha) ; 67(6): 811-824, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-1943173

ABSTRACT

The unique functionality of Akkermansia muciniphila in gut microbiota indicates it to be an indispensable microbe for human welfare. The importance of A. muciniphila lies in its potential to convert mucin into beneficial by-products, regulate intestinal homeostasis and maintain gut barrier integrity. It is also known to competitively inhibit other mucin-degrading bacteria and improve metabolic functions and immunity responses in the host. It finds a pivotal perspective in various diseases and their treatment. It has future as a promising probiotic, disease biomarker and therapeutic agent for chronic diseases. Disease-associated dysbiosis of A. muciniphila in the gut microbiome makes it a potential candidate as a biomarker for some diseases and can provide future theranostics by suggesting ways of diagnosis for the patients and best treatment method based on the screening results. Manipulation of A. muciniphila in gut microbiome may help in developing a novel personalized therapeutic action and can be a suitable next generation medicine. However, the actual pathway governing A. muciniphila interaction with hosts remains to be investigated. Also, due to the limited availability of products containing A. muciniphila, it is not exploited to its full potential. The present review aims at highlighting the potential of A. muciniphila in mucin degradation, contribution towards the gut health and host immunity and management of metabolic diseases such as obesity and type 2 diabetes, and respiratory diseases such as cystic fibrosis and COVID-19.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Humans , Dysbiosis/therapy , Verrucomicrobia/metabolism , Mucins/metabolism , Mucus
4.
Autophagy ; 18(10): 2350-2367, 2022 10.
Article in English | MEDLINE | ID: covidwho-1671990

ABSTRACT

Zaire ebolavirus (EBOV) causes a severe hemorrhagic fever in humans and non-human primates with high morbidity and mortality. EBOV infection is dependent on its structural glycoprotein (GP), but high levels of GP expression also trigger cell rounding, detachment, and downregulation of many surface molecules that is thought to contribute to its high pathogenicity. Thus, EBOV has evolved an RNA editing mechanism to reduce its GP expression and increase its fitness. We now report that the GP expression is also suppressed at the protein level in cells by protein disulfide isomerases (PDIs). Although PDIs promote oxidative protein folding by catalyzing correct disulfide formation in the endoplasmic reticulum (ER), PDIA3/ERp57 adversely triggered the GP misfolding by targeting GP cysteine residues and activated the unfolded protein response (UPR). Abnormally folded GP was targeted by ER-associated protein degradation (ERAD) machinery and, unexpectedly, was degraded via the macroautophagy/autophagy-lysosomal pathway, but not the proteasomal pathway. PDIA3 also decreased the GP expression from other ebolavirus species but increased the GP expression from Marburg virus (MARV), which is consistent with the observation that MARV-GP does not cause cell rounding and detachment, and MARV does not regulate its GP expression via RNA editing during infection. Furthermore, five other PDIs also had a similar inhibitory activity to EBOV-GP. Thus, PDIs negatively regulate ebolavirus glycoprotein expression, which balances the viral life cycle by maximizing their infection but minimizing their cellular effect. We suggest that ebolaviruses hijack the host protein folding and ERAD machinery to increase their fitness via reticulophagy during infection.Abbreviations: 3-MA: 3-methyladenine; 4-PBA: 4-phenylbutyrate; ACTB: ß-actin; ATF: activating transcription factor; ATG: autophagy-related; BafA1: bafilomycin A1; BDBV: Bundibugyo ebolavirus; CALR: calreticulin; CANX: calnexin; CHX: cycloheximide; CMA: chaperone-mediated autophagy; ConA: concanamycin A; CRISPR: clusters of regularly interspaced short palindromic repeats; Cas9: CRISPR-associated protein 9; dsRNA: double-stranded RNA; EBOV: Zaire ebolavirus; EDEM: ER degradation enhancing alpha-mannosidase like protein; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; Env: envelope glycoprotein; ER: endoplasmic reticulum; ERAD: ER-associated protein degradation; ERN1/IRE1: endoplasmic reticulum to nucleus signaling 1; GP: glycoprotein; HA: hemagglutinin; HDAC6: histone deacetylase 6; HMM: high-molecular-mass; HIV-1: human immunodeficiency virus type 1; HSPA5/BiP: heat shock protein family A (Hsp70) member 5; IAV: influenza A virus; IP: immunoprecipitation; KIF: kifenesine; Lac: lactacystin; LAMP: lysosomal associated membrane protein; MAN1B1/ERManI: mannosidase alpha class 1B member 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MARV: Marburg virus; MLD: mucin-like domain; NHK/SERPINA1: alpha1-antitrypsin variant null (Hong Kong); NTZ: nitazoxanide; PDI: protein disulfide isomerase; RAVV: Ravn virus; RESTV: Reston ebolavirus; SARS-CoV: severe acute respiratory syndrome coronavirus; SBOV: Sudan ebolavirus; sGP: soluble GP; SQSTM1/p62: sequestosome 1; ssGP: small soluble GP; TAFV: Taï Forest ebolavirus; TIZ: tizoxanide; TGN: thapsigargin; TLD: TXN (thioredoxin)-like domain; Ub: ubiquitin; UPR: unfolded protein response; VLP: virus-like particle; VSV: vesicular stomatitis virus; WB: Western blotting; WT: wild-type; XBP1: X-box binding protein 1.


Subject(s)
Autophagy , Ebolavirus , Actins/metabolism , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Protein 9/pharmacology , Calnexin/metabolism , Calreticulin/genetics , Calreticulin/metabolism , Calreticulin/pharmacology , Cycloheximide , Cysteine/metabolism , Disulfides , Endoplasmic Reticulum/metabolism , Glycoproteins/metabolism , Heat-Shock Proteins/metabolism , Hemagglutinins/metabolism , Hemagglutinins/pharmacology , Histone Deacetylase 6/genetics , Intercellular Signaling Peptides and Proteins , Lysosome-Associated Membrane Glycoproteins/metabolism , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Mucins/genetics , Mucins/metabolism , Mucins/pharmacology , Prokaryotic Initiation Factor-2/genetics , Prokaryotic Initiation Factor-2/metabolism , Prokaryotic Initiation Factor-2/pharmacology , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/pharmacology , Sequestosome-1 Protein/metabolism , Thapsigargin/metabolism , Thapsigargin/pharmacology , Thioredoxins/genetics , Thioredoxins/metabolism , Thioredoxins/pharmacology , Ubiquitins/metabolism , X-Box Binding Protein 1/metabolism , alpha-Mannosidase/genetics , alpha-Mannosidase/metabolism , alpha-Mannosidase/pharmacology
6.
Theranostics ; 11(13): 6193-6213, 2021.
Article in English | MEDLINE | ID: covidwho-1224320

ABSTRACT

Rationale: The pandemic caused by the novel coronavirus SARS-CoV-2 is advancing rapidly. In particular, the number of severe courses of the disease is still dramatically high. An efficient drug therapy that helps to improve significantly the fatal combination of damages in the airway epithelia, in the extensive pulmonary microvascularization and finally multiorgan failure, is missing. The physiological, inorganic polymer, polyphosphate (polyP) is a molecule which could prevent the initial phase of the virus life cycle, the attachment of the virus to the target cells, and improve the epithelial integrity as well as the mucus barrier. Results: Surprisingly, polyP matches perfectly with the cationic groove on the RBD. Subsequent binding studies disclosed that polyP, with a physiological chain length of 40 phosphate residues, abolishes the binding propensity of the RBD to the ACE2 receptor. In addition to this first mode of action of polyP, this polymer causes in epithelial cells an increased gene expression of the major mucins in the airways, of MUC5AC and MUC1, as well as a subsequent glycoprotein production. MUC5AC forms a gel-like mucus layer trapping inhaled particles which are then transported out of the airways, while MUC1 constitutes the periciliary liquid layer and supports ciliary beating. As a third mode of action, polyP undergoes enzymatic hydrolysis of the anhydride bonds in the airway system by alkaline phosphatase, releasing metabolic energy. Conclusions: This review summarizes the state of the art of the biotherapeutic potential of the polymer polyP and the findings from basic research and outlines future biomedical applications.


Subject(s)
COVID-19 Drug Treatment , Pandemics/prevention & control , Polyphosphates/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , COVID-19/epidemiology , COVID-19/transmission , COVID-19/virology , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Mice , Mucins/metabolism , Nanoparticles/chemistry , Polyphosphates/chemistry , Polyphosphates/therapeutic use , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Virus Attachment/drug effects
7.
Trends Microbiol ; 29(11): 983-992, 2021 11.
Article in English | MEDLINE | ID: covidwho-1187873

ABSTRACT

Efficient penetration of the mucus layer is needed for respiratory viruses to avoid mucociliary clearance prior to infection. Many respiratory viruses bind to glycans on the heavily glycosylated mucins that give mucus its gel-like characteristics. Influenza viruses, some paramyxoviruses, and coronaviruses avoid becoming trapped in the mucus by releasing themselves by means of their envelope-embedded enzymes that destroy glycan receptors. For efficient infection, receptor binding and destruction need to be in balance with the host receptor repertoire. Establishment in a novel host species requires resetting of the balance to adapt to the different glycan repertoire encountered. Growing understanding of species-specific mucosal glycosylation patterns and the dynamic interaction with respiratory viruses identifies the mucus layer as a major host-range determinant and barrier for zoonotic transfer.


Subject(s)
Host Specificity , Viruses , Glycosylation , Mucins/metabolism , Mucus/metabolism , Polysaccharides/metabolism , Viruses/metabolism
8.
Physiol Rep ; 9(1): e14701, 2021 01.
Article in English | MEDLINE | ID: covidwho-1000669

ABSTRACT

The Corona Virus Infectious Disease-19 (COVID-19) pandemic has played havoc on both the global health and economy. It is necessary to find a molecular signature that differentiates between low-risk and high-risk individuals. Pathogens, including viruses of the upper respiratory tract, utilize mucin proteins to enter into host cells. Mucins are critical components of innate immunity and also play important roles in infectious disease progression. Their expression is regulated by different cytokines during infection and inflammation. A comparison of mucin signatures between an asymptomatic versus symptomatic and between patients with mild versus severe symptoms could help identify other important proteins involved in the pathology of this new virus. Recent studies on the pathogenicity of the SARS-CoV-2 have found receptors that help its entry into the cells. In this review, we present an overview of how mucins are connected to the pathogenicity of the virus and propose that studying the glycome and mucin signature may lead to the development of a biomarker in predicting the susceptibility, progression, and response to therapy in COVID-19 patients.


Subject(s)
COVID-19 , Mucins/metabolism , SARS-CoV-2/pathogenicity , Animals , COVID-19/metabolism , COVID-19/virology , Humans , Transcriptome
9.
Life Sci ; 268: 118959, 2021 Mar 01.
Article in English | MEDLINE | ID: covidwho-988728

ABSTRACT

Cystic fibrosis (CF) is an autosomal recessive disease which involves the mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF involves in the inflammatory processes and is considered as a multisystem disorder that is not confined to lungs, but it also affects other vital organs that leads to numerous co-morbidities. The respiratory disorder in the CF results in mortality and morbidity which is characterized by series of serious events involving mucus hypersecretion, microbial infections, airways obstruction, inflammation, destruction of epithelium, tissue remodeling and terminal lung diseases. Mucins are the high molecular weight glycoproteins important for the viscoelastic properties of the mucus, play a significant role in the disease mechanisms. Determining the functional association between the CFTR and mucins might help to identify the putative target for specific therapeutic approach. In fact, furin enzyme which helps in the entry of novel COVID-19 virus into the cell, is upregulated in CF and this can also serve as a potential target for CF treatment. Moreover, the use of nano-formulations for CF treatment is an area of research being widely studied as they have also demonstrated promising outcomes. The in-depth knowledge of non-coding RNAs like miRNAs and lncRNAs and their functional association with CFTR gene expression and mutation can provide a different range of opportunity to identify the promising therapeutic approaches for CF.


Subject(s)
COVID-19/virology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/physiopathology , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/therapy , Gene Expression Regulation , Humans , MicroRNAs/genetics , Mucins/metabolism , Mutation , RNA, Long Noncoding/genetics , SARS-CoV-2/pathogenicity
10.
Nat Commun ; 11(1): 6319, 2020 12 09.
Article in English | MEDLINE | ID: covidwho-966313

ABSTRACT

The relationship of SARS-CoV-2 pulmonary infection and severity of disease is not fully understood. Here we show analysis of autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter-patient and intra-patient heterogeneity of pulmonary virus infection. There is a spectrum of high and low virus cases associated with duration of disease. High viral cases have high activation of interferon pathway genes and a predominant M1-like macrophage infiltrate. Low viral cases are more heterogeneous likely reflecting inherent patient differences in the evolution of host response, but there is consistent indication of pulmonary epithelial cell recovery based on napsin A immunohistochemistry and RNA expression of surfactant and mucin genes. Using a digital spatial profiling platform, we find the virus corresponds to distinct spatial expression of interferon response genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.


Subject(s)
COVID-19 , Host Microbial Interactions , Interferons/metabolism , Lung , Adult , Aged , Aged, 80 and over , Aspartic Acid Endopeptidases/metabolism , Autopsy , COVID-19/immunology , COVID-19/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Immunity , Immunohistochemistry , In Situ Hybridization , Interferons/genetics , Lung/pathology , Lung/virology , Macrophages/immunology , Male , Middle Aged , Mucins/genetics , Mucins/metabolism , Surface-Active Agents/metabolism , Transcriptome , Viral Load
11.
mBio ; 11(6)2020 11 12.
Article in English | MEDLINE | ID: covidwho-922531

ABSTRACT

Mucus plays a pivotal role in protecting the respiratory tract against microbial infections. It acts as a primary contact site to entrap microbes and facilitates their removal from the respiratory tract via the coordinated beating of motile cilia. The major components of airway mucus are heavily O-glycosylated mucin glycoproteins, divided into gel-forming mucins and transmembrane mucins. The gel-forming mucins MUC5AC and MUC5B are the primary structural components of airway mucus, and they enable efficient clearance of pathogens by mucociliary clearance. MUC5B is constitutively expressed in the healthy airway, whereas MUC5AC is upregulated in response to inflammatory challenge. MUC1, MUC4, and MUC16 are the three major transmembrane mucins of the respiratory tracts which prevent microbial invasion, can act as releasable decoy receptors, and activate intracellular signal transduction pathways. Pathogens have evolved virulence factors such as adhesins that facilitate interaction with specific mucins and mucin glycans, for example, terminal sialic acids. Mucin expression and glycosylation are dependent on the inflammatory state of the respiratory tract and are directly regulated by proinflammatory cytokines and microbial ligands. Gender and age also impact mucin glycosylation and expression through the female sex hormone estradiol and age-related downregulation of mucin production. Here, we discuss what is currently known about the role of respiratory mucins and their glycans during bacterial and viral infections of the airways and their relevance for the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Understanding the impact of microbe-mucin interaction in the respiratory tract could inspire the development of novel therapies to boost mucosal defense and combat respiratory infections.


Subject(s)
Glycoproteins/metabolism , Mucins/metabolism , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Bacterial Infections/metabolism , COVID-19/virology , Glycosylation , Humans , Mucin 5AC/metabolism , Mucin-1/metabolism , Mucin-5B/metabolism , Respiratory Tract Infections/prevention & control , SARS-CoV-2/pathogenicity , Virus Diseases/metabolism
12.
Cell Res ; 30(12): 1078-1087, 2020 12.
Article in English | MEDLINE | ID: covidwho-912896

ABSTRACT

Silent hypoxia has emerged as a unique feature of coronavirus disease 2019 (COVID-19). In this study, we show that mucins are accumulated in the bronchoalveolar lavage fluid (BALF) of COVID-19 patients and are upregulated in the lungs of severe respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected mice and macaques. We find that induction of either interferon (IFN)-ß or IFN-γ upon SARS-CoV-2 infection results in activation of aryl hydrocarbon receptor (AhR) signaling through an IDO-Kyn-dependent pathway, leading to transcriptional upregulation of the expression of mucins, both the secreted and membrane-bound, in alveolar epithelial cells. Consequently, accumulated alveolar mucus affects the blood-gas barrier, thus inducing hypoxia and diminishing lung capacity, which can be reversed by blocking AhR activity. These findings potentially explain the silent hypoxia formation in COVID-19 patients, and suggest a possible intervention strategy by targeting the AhR pathway.


Subject(s)
Interferons/metabolism , Mucus/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , COVID-19/pathology , COVID-19/virology , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Hypoxia , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Lung/metabolism , Lung/pathology , Macaca , Mice , Mice, Inbred ICR , Mice, Transgenic , Mucins/metabolism , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , Signal Transduction , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL