Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Mar Drugs ; 19(2)2021 Jan 27.
Article in English | MEDLINE | ID: covidwho-1969360

ABSTRACT

Inorganic polyphosphate (polyP) is a widely distributed polymer found from bacteria to animals, including marine species. This polymer exhibits morphogenetic as well as antiviral activity and releases metabolic energy after enzymatic hydrolysis also in human cells. In the pathogenesis of the coronavirus disease 2019 (COVID-19), the platelets are at the frontline of this syndrome. Platelets release a set of molecules, among them polyP. In addition, the production of airway mucus, the first line of body defense, is impaired in those patients. Therefore, in this study, amorphous nanoparticles of the magnesium salt of polyP (Mg-polyP-NP), matching the size of the coronavirus SARS-CoV-2, were prepared and loaded with the secondary plant metabolite quercetin or with dexamethasone to study their effects on the respiratory epithelium using human alveolar basal epithelial A549 cells as a model. The results revealed that both compounds embedded into the polyP nanoparticles significantly increased the steady-state-expression of the MUC5AC gene. This mucin species is the major mucus glycoprotein present in the secreted gel-forming mucus. The level of gene expression caused by quercetin or with dexamethasone, if caged into polyP NP, is significantly higher compared to the individual drugs alone. Both quercetin and dexamethasone did not impair the growth-supporting effect of polyP on A549 cells even at concentrations of quercetin which are cytotoxic for the cells. A possible mechanism of the effects of the two drugs together with polyP on mucin expression is proposed based on the scavenging of free oxygen species and the generation of ADP/ATP from the polyP, which is needed for the organization of the protective mucin-based mucus layer.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Dexamethasone/pharmacology , Mucin 5AC/biosynthesis , Mucin 5AC/drug effects , Quercetin/pharmacology , A549 Cells , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , COVID-19 , Dexamethasone/chemistry , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Humans , Magnesium/chemistry , Mucin 5AC/genetics , Mucins/biosynthesis , Mucins/chemistry , Nanoparticles , Particle Size , Plants/chemistry , Polyphosphates/chemistry , Quercetin/chemistry , Reactive Oxygen Species
2.
Biomater Sci ; 9(18): 6052-6063, 2021 Sep 14.
Article in English | MEDLINE | ID: covidwho-1287827

ABSTRACT

Conventional face masks to prevent SARS-CoV-2 transmission are mostly based on a passive filtration principle. Ideally, anti-COVID-19 masks should protect the carrier not only by size exclusion of virus aerosol particles, but also be able to capture and destroy or inactivate the virus. Here we present the proof-of-concept of a filter mat for such a mask, which actively attracts aerosol droplets and kills the virus. The electrospun mats are made of polycaprolactone (PCL) a hydrophilic, functionalizable and biodegradable polyester, into which inorganic polyphosphate (polyP) a physiological biocompatible, biodegradable and antivirally active polymer (chain length, ∼40 Pi units) has been integrated. A soluble Na-polyP as well as amorphous calcium polyP nanoparticles (Ca-polyP-NP) have been used. In this composition, the polyP component of the polyP-PCL mats is stable in aqueous protein-free environment, but capable of transforming into a gel-like coacervate upon contact with divalent cations and protein like mucin present in (virus containing) aerosol droplets. In addition, the Ca-polyP-NP are used as a carrier of tretinoin (all-trans retinoic acid) which blocks the function of the SARS-CoV-2 envelope (E) protein, an ion channel forming viroporin. The properties of this novel mask filter mats are as follows: First, to attract and to trap virus-like particles during the polyP coacervate formation induced in situ by aerosol droplets on the spun PCL fibers, as shown here by using SARS-CoV-2 mimicking fluorescent nanoparticles. Second, after disintegration the NP by the aerosol-mucus constituents, to release polyP that binds to and abolishes the function of the receptor binding domain of the viral spike protein. Third, to destroy the virus by releasing tretinoin, as shown by the disruption of virus-mimicking liposomes with the integrated recombinant viral viroporin. It is proposed that these properties, which are inducible (stimuli responsive), will allow the design of antiviral masks that are smart.


Subject(s)
COVID-19 , Nanoparticles , Humans , Masks , Polyphosphates , SARS-CoV-2
3.
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
4.
Cancers (Basel) ; 13(4)2021 Feb 11.
Article in English | MEDLINE | ID: covidwho-1090373

ABSTRACT

The anti-cancer antitumor antibiotic bleomycin(s) (BLM) induces athyminic sites in DNA after its activation, a process that results in strand splitting. Here, using A549 human lung cells or BEAS-2B cells lunc cells, we show that the cell toxicity of BLM can be suppressed by addition of inorganic polyphosphate (polyP), a physiological polymer that accumulates and is released from platelets. BLM at a concentration of 20 µg ml-1 causes a decrease in cell viability (by ~70%), accompanied by an increased DNA damage and chromatin expansion (by amazingly 6-fold). Importantly, the BLM-caused effects on cell growth and DNA integrity are substantially suppressed by polyP. In parallel, the enlargement of the nuclei/chromatin in BLM-treated cells (diameter, 20-25 µm) is normalized to ~12 µm after co-incubation of the cells with BLM and polyP. A sequential application of the drugs (BLM for 3 days, followed by an exposure to polyP) does not cause this normalization. During co-incubation of BLM with polyP the gene for the BLM hydrolase is upregulated. It is concluded that by upregulating this enzyme polyP prevents the toxic side effects of BLM. These data might also contribute to an application of BLM in COVID-19 patients, since polyP inhibits binding of SARS-CoV-2 to cellular ACE2.

5.
Mar Drugs ; 18(12)2020 Dec 14.
Article in English | MEDLINE | ID: covidwho-977761

ABSTRACT

The mucus layer of the nasopharynx and bronchial epithelium has a barrier function against inhaled pathogens such as the coronavirus SARS-CoV-2. We recently found that inorganic polyphosphate (polyP), a physiological, metabolic energy (ATP)-providing polymer released from blood platelets, blocks the binding of the receptor binding domain (RBD) to the cellular ACE2 receptor in vitro. PolyP is a marine natural product and is abundantly present in marine bacteria. Now, we have approached the in vivo situation by studying the effect of polyP on the human alveolar basal epithelial A549 cells in a mucus-like mucin environment. These cells express mucins as well as the ectoenzymes alkaline phosphatase (ALP) and adenylate kinase (ADK), which are involved in the extracellular production of ATP from polyP. Mucin, integrated into a collagen-based hydrogel, stimulated cell growth and attachment. The addition of polyP to the hydrogel significantly increased cell attachment and also the expression of the membrane-tethered mucin MUC1 and the secreted mucin MUC5AC. The increased synthesis of MUC1 was also confirmed by immunostaining. This morphogenetic effect of polyP was associated with a rise in extracellular ATP level. We conclude that the nontoxic and non-immunogenic polymer polyP could possibly also exert a protective effect against SARS-CoV-2-cell attachment; first, by stimulating the innate antiviral response by strengthening the mucin barrier with its antimicrobial proteins, and second, by inhibiting virus attachment to the cells, as deduced from the reduction in the strength of binding between the viral RBD and the cellular ACE2 receptor.


Subject(s)
Aquatic Organisms/metabolism , Biological Products/pharmacology , COVID-19/prevention & control , Polyphosphates/pharmacology , Respiratory Mucosa/drug effects , A549 Cells , Bacteria/metabolism , Biological Products/therapeutic use , COVID-19/virology , Humans , Immunity, Innate/drug effects , Mucin 5AC/metabolism , Mucin-1/metabolism , Polyphosphates/metabolism , Polyphosphates/therapeutic use , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Secondary Metabolism , Virus Attachment/drug effects
6.
Biomater Sci ; 8(23): 6603-6610, 2020 Dec 07.
Article in English | MEDLINE | ID: covidwho-968358

ABSTRACT

The effect of the polyanionic polymer of inorganic polyphosphate (polyP) involved in innate immunity on the binding of the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein to the cellular ACE2 receptor was studied. The RBD surface comprises a basic amino acid stretch of four arginine residues which interact with the physiological polyP (polyP40) and polyP3. Subsequently, the interaction of RBD with ACE2 is sensitively inhibited. After the chemical modification of arginine, an increased inhibition by polyP, at a 1 : 1 molar ratio (polyP : RBP), is measured already at 0.1 µg mL-1. Heparin was ineffective. The results suggest a potential therapeutic benefit of polyP against SARS-CoV-2 infection.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Polymers/pharmacology , Polyphosphates/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Antiviral Agents/chemistry , Binding Sites , Humans , Molecular Docking Simulation , Polyelectrolytes , Polymers/chemistry , Polyphosphates/chemistry , Protein Binding/drug effects , Spike Glycoprotein, Coronavirus/chemistry
7.
Biochem Pharmacol ; 182: 114215, 2020 12.
Article in English | MEDLINE | ID: covidwho-743871

ABSTRACT

Inorganic polyphosphate (polyP) is a morphogenetically active and metabolic energy-delivering physiological polymer that is released from blood platelets. Here, we show that polyP efficiently inhibits the binding of the envelope spike (S)-protein of the coronavirus SARS-CoV-2, the causative agent of COVID-19, to its host cell receptor ACE2 (angiotensin-converting enzyme 2). To stabilize polyP against the polyP-degrading alkaline phosphatase, the soluble polymer was encapsulated in silica/polyP nanoparticles. Applying a binding assay, soluble Na-polyP (sizes of 40 Pi and of 3 Pi units) as well as silica-nanoparticle-associated polyP significantly inhibit the interaction of the S-protein with ACE2 at a concentration of 1 µg/mL, close to the level present in blood. This inhibition is attributed to an interaction of polyP with a basic amino acid stretch on the surface of the receptor binding domain of S-protein. PolyP retains its activity in a flushing solution, opening a new strategy for the prevention and treatment of SARS-CoV-2 infection in the oropharyngeal cavity. The data suggest that supplementation of polyP might contribute to a strengthening of the human innate immunity system in compromised, thrombocytopenic COVID-19 patients.


Subject(s)
Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Antiviral Agents/pharmacology , Polyphosphates/pharmacology , Receptors, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Nanoparticles/therapeutic use , Polyphosphates/chemistry , Protein Binding/drug effects , Receptors, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL