Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Front Pharmacol ; 14: 1087850, 2023.
Article in English | MEDLINE | ID: covidwho-2323285

ABSTRACT

Acute respiratory viral infections (ARVI) are the most common illnesses worldwide. In some instances, mild cases of ARVI progress to hyperinflammatory responses, which are damaging to pulmonary tissue and requiring intensive care. Here we summarize available information on preclinical and clinical effects of XC221GI (1-[2-(1-methyl imidazole-4-yl)-ethyl]perhydroazin-2,6-dione), an oral drug with a favorable safety profile that has been tested in animal models of influenza, respiratory syncytial virus, highly pathogenic coronavirus strains and other acute viral upper respiratory infections. XC221GI is capable of controlling IFN-gamma-driven inflammation as it is evident from the suppression of the production of soluble cytokines and chemokines, including IL-6, IL-8, CXCL10, CXCL9 and CXCL11 as well as a decrease in migration of neutrophils into the pulmonary tissue. An excellent safety profile of XC221GI, which is not metabolized by the liver, and its significant anti-inflammatory effects indicate utility of this compound in abating conversion of ambulatory cases of respiratory infections into the cases with aggravated presentation that require hospitalization. This drug is especially useful when rapid molecular assays determining viral species are impractical, or when direct antiviral drugs are not available. Moreover, XC221GI may be combined with direct antiviral drugs to enhance their therapeutic effects.

2.
Front Immunol ; 14: 1030879, 2023.
Article in English | MEDLINE | ID: covidwho-2309368

ABSTRACT

Introduction: There is an unmet medical need for effective anti-inflammatory agents for the treatment of acute and post-acute lung inflammation caused by respiratory viruses. The semi-synthetic polysaccharide, Pentosan polysulfate sodium (PPS), an inhibitor of NF-kB activation, was investigated for its systemic and local anti-inflammatory effects in a mouse model of influenza virus A/PR8/1934 (PR8 strain) mediated infection. Methods: Immunocompetent C57BL/6J mice were infected intranasally with a sublethal dose of PR8 and treated subcutaneously with 3 or 6 mg/kg PPS or vehicle. Disease was monitored and tissues were collected at the acute (8 days post-infection; dpi) or post-acute (21 dpi) phase of disease to assess the effect of PPS on PR8-induced pathology. Results: In the acute phase of PR8 infection, PPS treatment was associated with a reduction in weight loss and improvement in oxygen saturation when compared to vehicle-treated mice. Associated with these clinical improvements, PPS treatment showed a significant retention in the numbers of protective SiglecF+ resident alveolar macrophages, despite uneventful changes in pulmonary leukocyte infiltrates assessed by flow cytometry. PPS treatment in PR8- infected mice showed significant reductions systemically but not locally of the inflammatory molecules, IL-6, IFN-g, TNF-a, IL-12p70 and CCL2. In the post-acute phase of infection, PPS demonstrated a reduction in the pulmonary fibrotic biomarkers, sICAM-1 and complement factor C5b9. Discussion: The systemic and local anti-inflammatory actions of PPS may regulate acute and post-acute pulmonary inflammation and tissue remodeling mediated by PR8 infection, which warrants further investigation.


Subject(s)
Influenzavirus A , Pneumonia , Mice , Animals , Pentosan Sulfuric Polyester/pharmacology , Pentosan Sulfuric Polyester/therapeutic use , Mice, Inbred C57BL , Pneumonia/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal
3.
mBio ; 14(2): e0313722, 2023 04 25.
Article in English | MEDLINE | ID: covidwho-2263060

ABSTRACT

Acute respiratory distress syndrome (ARDS) is triggered by a variety of insults, including bacterial and viral infections, and this leads to high mortality. While the role of the aryl hydrocarbon receptor (AhR) in mucosal immunity is being increasingly recognized, its function during ARDS is unclear. In the current study, we investigated the role of AhR in LPS-induced ARDS. AhR ligand, indole-3-carbinol (I3C), attenuated ARDS which was associated with a decrease in CD4+ RORγt +IL-17a+IL-22+ pathogenic Th17 cells, but not CD4+RORγt +IL-17a+IL-22- homeostatic Th 17 cells, in the lungs. AhR activation also led to a significant increase in CD4+IL-17a-IL-22+ Th22 cells. I3C-mediated Th22 cell expansion was dependent on the AhR expression on RORγt+ cells. AhR activation downregulated miR-29b-2-5p in immune cells from the lungs, which in turn downregulated RORc expression and upregulated IL-22. Collectively, the current study suggests that AhR activation can attenuate ARDS and may serve as a therapeutic modality by which to treat this complex disorder. IMPORTANCE Acute respiratory distress syndrome (ARDS) is a type of respiratory failure that is triggered by a variety of bacterial and viral infections, including the coronavirus SARS-CoV2. ARDS is associated with a hyperimmune response in the lungs that which is challenging to treat. Because of this difficulty, approximately 40% of patients with ARDS die. Thus, it is critical to understand the nature of the immune response that is functional in the lungs during ARDS as well as approaches by which to attenuate it. AhR is a transcription factor that is activated by a variety of endogenous and exogenous environmental chemicals as well as bacterial metabolites. While AhR has been shown to regulate inflammation, its role in ARDS is unclear. In the current study, we provide evidence that AhR activation can attenuate LPS-mediated ARDS through the activation of Th22 cells in the lungs, which are regulated through miR-29b-2-5p. Thus, AhR can be targeted to attenuate ARDS.


Subject(s)
MicroRNAs , Receptors, Aryl Hydrocarbon , Respiratory Distress Syndrome , Humans , Interleukin-17 , Lipopolysaccharides , Lung/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Respiratory Distress Syndrome/pathology , RNA, Viral , SARS-CoV-2/metabolism , Th17 Cells
4.
Front Neurol ; 14: 1052811, 2023.
Article in English | MEDLINE | ID: covidwho-2271738

ABSTRACT

Substance P (SP) has been a great interest for scientists due to its unique properties and involvement in various physiological and pathological phenomenon. It took almost a century for the current understanding of this peptide so far. Its role in brain and gut were initially discussed and later on it was widely studied and observed in cardiovascular system, asthma, traumatic brain injury, immune response, vasodilation, behavior, inflammation, arthritis, cancer, airway hyper responsiveness and respiratory disorders. Involvement of SP in sudden perinatal death and COVID-19 has also been discussed which shed light on its vital role in respiratory rhythm regulation and initiation of cytokine storming in COVID-19. This article will provide a comprehensive overview of the researches done to understand the basic functions and involvement of SP in different processes of cell and its association with various diseases. This article describes the historical and scientific journey of SP from its discovery until today, including its future perspectives.

5.
Vaccines (Basel) ; 11(1)2022 Dec 21.
Article in English | MEDLINE | ID: covidwho-2236640

ABSTRACT

Following the breakthrough of numerous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants in recent months and the incomplete efficiency of the currently available vaccines, development of more effective vaccines is desirable. Non-integrative, non-cytopathic and non-inflammatory lentiviral vectors elicit sterilizing prophylaxis against SARS-CoV-2 in preclinical animal models and are particularly suitable for mucosal vaccination, which is acknowledged as the most effective in reducing viral transmission. Here, we demonstrate that a single intranasal administration of a vaccinal lentiviral vector encoding a stabilized form of the original SARS-CoV-2 Spike glycoprotein induces full-lung protection of respiratory tracts and strongly reduces pulmonary inflammation in the susceptible Syrian golden hamster model against the prototype SARS-CoV-2. In addition, we show that a lentiviral vector encoding stabilized Spike of SARS-CoV-2 Beta variant (LV::SBeta-2P) prevents pathology and reduces infectious viral loads in lungs and nasal turbinates following inoculation with the SARS-CoV-2 Omicron variant. Importantly, an intranasal boost with LV::SBeta-2P improves cross-seroneutralization much better in LV::SBeta-2P-primed hamsters than in their counterparts primed with an LV-encoding Spike from the ancestral SARS-CoV-2. These results strongly suggest that an immune imprint with the original Spike sequence has a negative impact on cross-protection against new variants. Our results tackle the issue of vaccine effectiveness in people who have already been vaccinated and have vanished immunity and indicate the efficiency of LV-based intranasal vaccination, either as a single dose or as booster.

6.
Front Med (Lausanne) ; 9: 1072056, 2022.
Article in English | MEDLINE | ID: covidwho-2229520

ABSTRACT

Objective: The multi-systemic inflammation as a result of COVID-19 can persevere long after the initial symptoms of the illness have subsided. These effects are referred to as Long-COVID. Our research focused on the contribution of the Spike protein S1 subunit of SARS-CoV-2 (Spike S1) on the lung inflammation mediated by NLRP3 inflammasome machinery and the cytokine releases, interleukin 6 (IL-6), IL-1beta, and IL-18, in lung epithelial cells. This study has attempted to identify the naturally- occurring agents that act against inflammation-related long-COVID. The seed meal of Perilla frutescens (P. frutescens), which contains two major dietary polyphenols (rosmarinic acid and luteolin), has been reported to exhibit anti-inflammation activities. Therefore, we have established the ethyl acetate fraction of P. frutescens seed meal (PFEA) and determined its anti-inflammatory effects on Spike S1 exposure in A549 lung cells. Methods: PFEA was established using solvent-partitioned extraction. Rosmarinic acid (Ra) and luteolin (Lu) in PFEA were identified using the HPLC technique. The inhibitory effects of PFEA and its active compounds against Spike S1-induced inflammatory response in A549 cells were determined by RT-PCR and ELISA. The mechanistic study of anti-inflammatory properties of PFEA and Lu were determined using western blot technique. Results: PFEA was found to contain Ra (388.70 ± 11.12 mg/g extract) and Lu (248.82 ± 12.34 mg/g extract) as its major polyphenols. Accordingly, A549 lung cells were pre-treated with PFEA (12.5-100 µg/mL) and its two major compounds (2.5-20 µg/mL) prior to the Spike S1 exposure at 100 ng/mL. PFEA dose-dependently exhibited anti-inflammatory properties upon Spike S1-exposed A549 cells through IL-6, IL-1ß, IL-18, and NLRP3 gene suppressions, as well as IL-6, IL-1ß, and IL-18 cytokine releases with statistical significance (p < 0.05). Importantly, Lu possesses superior anti-inflammatory properties when compared with Ra (p < 0.01). Mechanistically, PFEA and Lu effectively attenuated a Spike S1-induced inflammatory response through downregulation of the JAK1/STAT3-inflammasome-dependent inflammatory pathway as evidenced by the downregulation of NLRP3, ASC, and cleaved-caspase-1 of the NLRP3 inflammasome components and by modulating the phosphorylation of JAK1 and STAT3 proteins (p < 0.05). Conclusion: The findings suggested that luteolin and PFEA can modulate the signaling cascades that regulate Spike S1-induced lung inflammation during the incidence of Long-COVID. Consequently, luteolin and P. frutescens may be introduced as potential candidates in the preventive therapeutic strategy for inflammation-related post-acute sequelae of COVID-19.

7.
Int Immunopharmacol ; 115: 109701, 2023 Feb.
Article in English | MEDLINE | ID: covidwho-2179731

ABSTRACT

Acute respiratory distress syndrome (ARDS) is associated with severe lung inflammation, edema, hypoxia, and high vascular permeability. The COVID-19-associated pandemic ARDS caused by SARS-CoV-2 has created dire global conditions and has been highly contagious. Chronic inflammatory disease enhances cancer cell proliferation, progression, and invasion. We investigated how acute lung inflammation activates the tumor microenvironment and enhances lung metastasis in LPS induced in vitro and in vivo models. Respiratory illness is mainly caused by cytokine storm, which further influences oxidative and nitrosative stress. The LPS-induced inflammatory cytokines made the conditions suitable for the tumor microenvironment in the lungs. In the present study, we observed that LPS induced the cytokine storm and promoted lung inflammation via BRD4, which further caused the nuclear translocation of p65 NF-κB and STAT3. The transcriptional activation additionally triggers the tumor microenvironment and lung metastasis. Thus, BRD4-regulated p65 and STAT3 transcriptional activity in ARDS enhances lung tumor metastasis. Moreover, LPS-induced ARDS might promote the tumor microenvironment and increase cancer metastasis into the lungs. Collectively, BRD4 plays a vital role in inflammation-mediated tumor metastasis and is found to be a diagnostic and molecular target in inflammation-associated cancers.


Subject(s)
COVID-19 , Lung Neoplasms , Pneumonia , Respiratory Distress Syndrome , Humans , Nuclear Proteins/genetics , Lipopolysaccharides/pharmacology , Tumor Microenvironment , Cytokine Release Syndrome , SARS-CoV-2 , Transcription Factors/genetics , Lung/pathology , Respiratory Distress Syndrome/chemically induced , Pneumonia/chemically induced , Inflammation , Cell Cycle Proteins/genetics
8.
3rd IEEE Global Conference for Advancement in Technology, GCAT 2022 ; 2022.
Article in English | Scopus | ID: covidwho-2191781

ABSTRACT

In this work, an attempt has been made to develop an Internet-of-Medical-Things (IoMT) based tracking system for cardiopulmonary disease affected patients. COVID-19 pandemic has caused a wide range of illness and organ failure to human beings around the world. Severe infection could alter the heart function and result in damage of the cardiac muscles. The disease could cause lung inflammation, pneumonia, severe acute respiratory syndrome and sometimes resulting in death. To monitor the wellness of heart and lungs of patients recovering from COVID-19, an efficient wearable health tracking system is required. The proposed system consists of a sensor to monitor heart rate and Oxygen Saturation (SpO2) of an individual. The sensor data is sent to an Arduino UNO Microcontroller. The normal range of heart rate and oxygen saturation are set as threshold in the control unit. If the heart rate and SpO2 levels are above or below the threshold, the control module will send an alert to the nearby nursing station and provide treatment to the patient. The patient data is continuously recorded and stored in the hospital's cloud as electronic health record. The prototype was tested in individuals in relaxed state and after performing tasks such as stair climbing. Results indicate that the proposed system could accurately track the status of the heart function along with oxygen saturation levels. Hence, this could be used as a cardiopulmonary health tracking device thereby assisting the clinicians in providing appropriate critical care services to the infected patients. © 2022 IEEE.

9.
Nutrients ; 14(23)2022 Nov 25.
Article in English | MEDLINE | ID: covidwho-2123778

ABSTRACT

In the wake of the COVID-19 pandemic, lung disorders have become a major health concern for humans. Allergic asthma is the most prevalent form of asthma, and its treatments target the inflammation process. Despite significant developments in the diagnosis and management of allergic asthma, side effects are a major concern. Additionally, its extreme heterogeneity impedes the efficacy of the majority of treatments. Thus, newer, safer therapeutic substances, such as natural products, are desired. Citrus junos Tanaka has traditionally been utilized as an anti-inflammatory, sedative, antipyretic, and antitoxic substance. In this study, the protective effects of Citrus junos Tanaka peel extract (B215) against lung inflammation were examined, and efforts were made to understand the underlying protective mechanism using an HDM-induced lung inflammation murine model. The administration of B215 reduced immune cell infiltration in the lungs, plasma IgE levels, airway resistance, mucus hypersecretions, and cytokine production. These favorable effects alleviated HDM-induced lung inflammation by modulating the NF-κB signaling pathway. Hence, B215 might be a promising functional food to treat lung inflammation without adverse effects.


Subject(s)
Asthma , COVID-19 , Citrus , Pneumonia , Mice , Humans , Animals , Pandemics , Disease Models, Animal , COVID-19/metabolism , Lung , Pneumonia/drug therapy , Pneumonia/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/metabolism , Immunity
10.
Clin Immunol ; 244: 109093, 2022 11.
Article in English | MEDLINE | ID: covidwho-2049018

ABSTRACT

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Emerging evidence indicates that the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome is activated, which results in a cytokine storm at the late stage of COVID-19. Autophagy regulation is involved in the infection and replication of SARS-CoV-2 at the early stage and the inhibition of NLRP3 inflammasome-mediated lung inflammation at the late stage of COVID-19. Here, we discuss the autophagy regulation at different stages of COVID-19. Specifically, we highlight the therapeutic potential of autophagy activators in COVID-19 by inhibiting the NLRP3 inflammasome, thereby avoiding the cytokine storm. We hope this review provides enlightenment for the use of autophagy activators targeting the inhibition of the NLRP3 inflammasome, specifically the combinational therapy of autophagy modulators with the inhibitors of the NLRP3 inflammasome, antiviral drugs, or anti-inflammatory drugs in the fight against COVID-19.


Subject(s)
COVID-19 , Pneumonia , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/pharmacology , Autophagy , Cytokine Release Syndrome , Humans , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , SARS-CoV-2
11.
Pharmacol Rep ; 74(6): 1315-1325, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-1982429

ABSTRACT

BACKGROUND: COVID-19, the disease caused by SARS-CoV-2 virus infection, has been a major public health problem worldwide in the last 2 years. SARS-CoV-2-dependent activation of innate immune receptors contributes to the strong local and systemic inflammatory reaction associated with rapid disease evolution. The receptor-binding domain (RBD) of Spike (S) viral protein (S-RBD) is essential for virus infection and its interacting molecules in target cells are still under identification. On the other hand, the search for accessible natural molecules with potential therapeutic use has been intense and remains an active field of investigation. METHODS: C57BL6/J (control) and Toll-like receptor (TLR) 4-deficient (Lps del) mice were nebulized with recombinant S-RBD. Tumor Necrosis Factor-alpha (TNF-α) and Interleukin (IL)-6 production in bronchoalveolar lavages (BALs) was determined by enzyme-linked immunosorbent assay (ELISA). Lung-infiltrating cells recovered in BALs were quantified by hematoxylin-eosin (H&E) stain. In selected groups of animals, the natural compound Jacareubin or dexamethasone were intraperitoneally (ip) administered 2 hours before nebulization. RESULTS: A rapid lung production of TNF-α and IL-6 and cell infiltration was induced by S-RBD nebulization in control but not in Lps del mice. Pre-treatment with Jacareubin or dexamethasone prevented S-RBD-induced TNF-α and IL-6 secretion in BALs from control animals. CONCLUSIONS: S-RBD domain promotes lung TNF-α and IL-6 production in a TLR4-dependent fashion in C57BL6/J mice. Xanthone Jacareubin possesses potential anti-COVID-19 properties that, together with the previously tested anti-inflammatory activity, safety, and tolerance, make it a valuable drug to be further investigated for the treatment of cytokine production caused by SARS-CoV-2 infection.

12.
Front Toxicol ; 4: 840606, 2022.
Article in English | MEDLINE | ID: covidwho-1933930

ABSTRACT

The evaluation of inhalation toxicity, drug safety and efficacy assessment, as well as the investigation of complex disease pathomechanisms, are increasingly relying on in vitro lung models. This is due to the progressive shift towards human-based systems for more predictive and translational research. While several cellular models are currently available for the upper airways, modelling the distal alveolar region poses several constraints that make the standardization of reliable alveolar in vitro models relatively difficult. In this work, we present a new and reproducible alveolar in vitro model, that combines a human derived immortalized alveolar epithelial cell line (AXiAEC) and organ-on-chip technology mimicking the lung alveolar biophysical environment (AXlung-on-chip). The latter mimics key features of the in vivo alveolar milieu: breathing-like 3D cyclic stretch (10% linear strain, 0.2 Hz frequency) and an ultrathin, porous and elastic membrane. AXiAECs cultured on-chip were characterized for their alveolar epithelial cell markers by gene and protein expression. Cell barrier properties were examined by TER (Transbarrier Electrical Resistance) measurement and tight junction formation. To establish a physiological model for the distal lung, AXiAECs were cultured for long-term at air-liquid interface (ALI) on-chip. To this end, different stages of alveolar damage including inflammation (via exposure to bacterial lipopolysaccharide) and the response to a profibrotic mediator (via exposure to Transforming growth factor ß1) were analyzed. In addition, the expression of relevant host cell factors involved in SARS-CoV-2 infection was investigated to evaluate its potential application for COVID-19 studies. This study shows that AXiAECs cultured on the AXlung-on-chip exhibit an enhanced in vivo-like alveolar character which is reflected into: 1) Alveolar type 1 (AT1) and 2 (AT2) cell specific phenotypes, 2) tight barrier formation (with TER above 1,000 Ω cm2) and 3) reproducible long-term preservation of alveolar characteristics in nearly physiological conditions (co-culture, breathing, ALI). To the best of our knowledge, this is the first time that a primary derived alveolar epithelial cell line on-chip representing both AT1 and AT2 characteristics is reported. This distal lung model thereby represents a valuable in vitro tool to study inhalation toxicity, test safety and efficacy of drug compounds and characterization of xenobiotics.

13.
Front Immunol ; 13: 875546, 2022.
Article in English | MEDLINE | ID: covidwho-1933655

ABSTRACT

Cytokine storm refers to the dysregulated production of inflammatory mediators leading to hyperinflammation. They are often detrimental, and worsen the severity of COVID-19 and other infectious or inflammatory diseases. Cannabinoids are known to have anti-inflammatory effects but their possible therapeutic value on cytokine storms has not been fully elucidated. In vivo and ex vivo studies were carried out to investigate the effects of high-THC and high-CBD extracts on cytokine production in immune cells. Significant differences between the extracts were observed. Subsequent experiments focusing on a specific high CBD extract (CBD-X) showed significant reductions in pro-inflammatory cytokines in human-derived PBMCs, neutrophils and T cells. In vivo mouse studies, using a systemically inflamed mouse model, showed reductions in pro-inflammatory cytokines TNFα and IL-1ß and a concurrent increase in the anti-inflammatory cytokine IL-10 in response to CBD-X extract treatment. Lung inflammation, as in severe COVID-19 disease, is characterized by increased T-cell homing to the lungs. Our investigation revealed that CBD-X extract impaired T-cell migration induced by the chemoattractant SDF1. In addition, the phosphorylation levels of T cell receptor (TCR) signaling proteins Lck and Zap70 were significantly reduced, demonstrating an inhibitory effect on the early events downstream to TCR activation. In a lung inflamed mouse model, we observed a reduction in leukocytes including neutrophil migration to the lungs and decreased levels of IL-1ß, MCP-1, IL-6 and TNFα, in response to the administration of the high-CBD extract. The results presented in this work offer that certain high-CBD extract has a high potential in the management of pathological conditions, in which the secretion of cytokines is dysregulated, as it is in severe COVID-19 disease or other infectious or inflammatory diseases.


Subject(s)
COVID-19 Drug Treatment , Cytokine Release Syndrome , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cytokine Release Syndrome/drug therapy , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Lung/metabolism , Mice , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Tumor Necrosis Factor-alpha
14.
Viruses ; 14(6)2022 05 24.
Article in English | MEDLINE | ID: covidwho-1911604

ABSTRACT

Without sufficient herd immunity through either vaccination or natural infection, the coronavirus disease 2019 pandemic is unlikely to be controlled. Waning immunity with the currently approved vaccines suggests the need to evaluate vaccines causing the induction of long-term responses. Here, we report the immunogenicity and efficacy of our adjuvanted single-dose Rabies-vectored SARS-CoV-2 S1 vaccine, CORAVAX, in hamsters. CORAVAX induces high SARS-CoV-2 S1-specific and virus-neutralizing antibodies (VNAs) that prevent weight loss, viral loads, disease, lung inflammation, and the cytokine storm in hamsters. We also observed high Rabies VNA titers. In summary, CORAVAX is a promising dual-antigen vaccine candidate for clinical evaluation against SARS-CoV-2 and Rabies virus.


Subject(s)
COVID-19 , Rabies Vaccines , Rabies virus , Rabies , Viral Vaccines , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Cricetinae , Humans , Rabies/prevention & control , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus
15.
Environ Sci Technol ; 56(7): 4272-4281, 2022 04 05.
Article in English | MEDLINE | ID: covidwho-1764112

ABSTRACT

Global spread of coronavirus disease-19 (COVID-19) is placing an unprecedented pressure on the environment and health. In this study, a new perspective is proposed to assess the inhalation bioaccessibility of polycyclic aromatic hydrocarbons (PAHs) in PM2.5 for people with various lung health conditions. In vitro bioaccessibility (IVBA) was measured using modified epithelial lung fluids simulating the extracellular environment of patients with severe and mild lung inflammation. The average PAH IVBA in PM2.5 of 24.5 ± 4.52% under healthy conditions increased (p = 0.06) to 28.6 ± 3.17% and significantly (p < 0.05) to 32.3 ± 5.32% under mild and severe lung inflammation conditions. A mechanistic study showed that lung inflammation decreased the critical micelle concentrations of main pulmonary surfactants (i.e., from 67.8 (for dipalmitoyl phosphatidylcholine) and 53.3 mg/L (for bovine serum albumin) to 44.5 mg/L) and promoted the formation of micelles, which enhanced the solubilization and competitive desorption of PAHs from PM2.5 in the lung fluids. In addition, risk assessment considering different IVBA values suggested that PAH contamination levels in PM2.5, which were safe for healthy people, may not be acceptable for patients with lung inflammation. Because of the large number of COVID-19 infections, and the fact that some survivors of COVID-19 were observed to still show symptoms of interstitial lung inflammation, the finding here can provide important implications for both the scientific community and policy makers in addressing health risk and air pollution control during the COVID-19 outbreak.


Subject(s)
Air Pollutants , Air Pollution , COVID-19 , Polycyclic Aromatic Hydrocarbons , Air Pollutants/analysis , COVID-19/epidemiology , China , Environmental Monitoring , Humans , Lung , Particulate Matter/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Risk Assessment
16.
J Control Release ; 346: 421-433, 2022 06.
Article in English | MEDLINE | ID: covidwho-1763813

ABSTRACT

Acute Respiratory Distress Syndrome (ARDS), associated with Covid-19 infections, is characterized by diffuse lung damage, inflammation and alveolar collapse that impairs gas exchange, leading to hypoxemia and patient' mortality rates above 40%. Here, we describe the development and assessment of 100-nm liposomes that are tailored for pulmonary delivery for treating ARDS, as a model for lung diseases. The liposomal lipid composition (primarily DPPC) was optimized to mimic the lung surfactant composition, and the drug loading process of both methylprednisolone (MPS), a steroid, and N-acetyl cysteine (NAC), a mucolytic agent, reached an encapsulation efficiency of 98% and 92%, respectively. In vitro, treating lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages with the liposomes decreased TNFα and nitric oxide (NO) secretion, while NAC increased the penetration of nanoparticles through the mucus. In vivo, we used LPS-induced lung inflammation model to assess the accumulation and therapeutic efficacy of the liposomes in C57BL/6 mice, either by intravenous (IV), endotracheal (ET) or IV plus ET nanoparticles administrations. Using both administration methods, liposomes exhibited an increased accumulation profile in the inflamed lungs over 48 h. Interestingly, while IV-administrated liposomes distributed widely throughout the lung, ET liposomes were present in lungs parenchyma but were not detected at some distal regions of the lungs, possibly due to imperfect airflow regimes. Twenty hours after the different treatments, lungs were assessed for markers of inflammation. We found that the nanoparticle treatment had a superior therapeutic effect compared to free drugs in treating ARDS, reducing inflammation and TNFα, IL-6 and IL-1ß cytokine secretion in bronchoalveolar lavage (BAL), and that the combined treatment, delivering nanoparticles IV and ET simultaneously, had the best outcome of all treatments. Interestingly, also the DPPC lipid component alone played a therapeutic role in reducing inflammatory markers in the lungs. Collectively, we show that therapeutic nanoparticles accumulate in inflamed lungs holding potential for treating lung disorders. SIGNIFICANCE: In this study we compare intravenous versus intratracheal delivery of nanoparticles for treating lung disorders, specifically, acute respiratory distress syndrome (ARDS). By co-loading two medications into lipid nanoparticles, we were able to reduce both inflammation and mucus secretion in the inflamed lungs. Both modes of delivery resulted in high nanoparticle accumulation in the lungs, intravenously administered nanoparticles reached lung endothelial while endotracheal delivery reached lung epithelial. Combining both delivery approaches simultaneously provided the best ARDS treatment outcome.


Subject(s)
COVID-19 , Lung Diseases , Respiratory Distress Syndrome , Acetylcysteine/pharmacology , Animals , Humans , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Liposomes/therapeutic use , Lung , Mice , Mice, Inbred C57BL , Nanoparticles , Respiratory Distress Syndrome/drug therapy , Tumor Necrosis Factor-alpha
17.
Balneo and Prm Research Journal ; 12(4):323-326, 2021.
Article in English | Web of Science | ID: covidwho-1689787

ABSTRACT

Introduction. An unprecedented public health crisis has been triggered worldwide by SARS-CoV-2's high contagiosity and it's mortality rates of 1-5%. Although the majority of COVID-19 cases have a good outcome, there is a small percentage that develop severe pneumonia and citokine storm and may be in the need of mechanical ventilation. Methods. Identifying the exact drivers of the excessive inflammation and the biomarkers that can predict a hyperinflammatory response to SARS-CoV-2 would be extremly helpful in finding efficient anti-inflammatory interventions that may stop the progression to acute respiratory distress syndrome (ARDS). Results. In the search for such interventions we have identified the promising effect of low level LASER therapy (LLLT) on lung inflammation from COVID-19 pneumonia. Due to its well known anti-inflammatory effect and modulatory activity on immune cells, laser therapy may be able to decrease lung and systemic inflammation without affecting lung function in acute lung lesions, relieve respiratory symptoms, normalize respiratory function and stimulate the healing process of lung tissue. The recovery time may also be significantly shortened and all blood, immunological and radiological parameters may improve. Conclusions. This findings need further confirmation from clinical trials but we are hopeful for their contribution on the global battle against COVID-19 pandemic.

18.
Methods Cell Biol ; 168: 329-341, 2022.
Article in English | MEDLINE | ID: covidwho-1628694

ABSTRACT

As more infectious viruses emerge that result in respiratory illness, there is a significant need to standardize airway harvests and maximize data acquisition. Animal models of respiratory viral infections have been outlined to allow for the analysis of the host immune response and viral pathogenesis kinetics. This chapter outlines two separate tissue harvest protocols following the intranasal infection of mice to investigate both the host immune response and viral pathogenesis. These protocols combine standard laboratory techniques for the analysis of the samples, making it easily integrable for labs without the need for specialized training. In offering two separate yet parallel tissue collection techniques, investigators can ultimately decide which technique will yield the best data for their particular research questions and can maximize data from each animal study.


Subject(s)
Pneumonia , Viruses , Animals , Immunity , Mice
19.
Respir Res ; 23(1): 25, 2022 Feb 10.
Article in English | MEDLINE | ID: covidwho-1677511

ABSTRACT

BACKGROUND: Pulmonary hyperinflammation is a key event with SARS-CoV-2 infection. Acute respiratory distress syndrome (ARDS) that often accompanies COVID-19 appears to have worse outcomes than ARDS from other causes. To date, numerous lung histological studies in cases of COVID-19 have shown extensive inflammation and injury, but the extent to which these are a COVID-19 specific, or are an ARDS and/or mechanical ventilation (MV) related phenomenon is not clear. Furthermore, while lung hyperinflammation with ARDS (COVID-19 or from other causes) has been well studied, there is scarce documentation of vascular inflammation in COVID-19 lungs. METHODS: Lung sections from 8 COVID-19 affected and 11 non-COVID-19 subjects, of which 8 were acute respiratory disease syndrome (ARDS) affected (non-COVID-19 ARDS) and 3 were from subjects with non-respiratory diseases (non-COVID-19 non-ARDS) were H&E stained to ascertain histopathological features. Inflammation along the vessel wall was also monitored by expression of NLRP3 and caspase 1. RESULTS: In lungs from COVID-19 affected subjects, vascular changes in the form of microthrombi in small vessels, arterial thrombosis, and organization were extensive as compared to lungs from non-COVID-19 (i.e., non-COVID-19 ARDS and non-COVID-19 non-ARDS) affected subjects. The expression of NLRP3 pathway components was higher in lungs from COVID-19 ARDS subjects as compared to non-COVID-19 non-ARDS cases. No differences were observed between COVID-19 ARDS and non-COVID-19 ARDS lungs. CONCLUSION: Vascular changes as well as NLRP3 inflammasome pathway activation were not different between COVID-19 and non-COVID-19 ARDS suggesting that these responses are not a COVID-19 specific phenomenon and are possibly more related to respiratory distress and associated strategies (such as MV) for treatment.


Subject(s)
Blood Vessels/immunology , COVID-19/immunology , Inflammasomes/analysis , Lung/blood supply , NLR Family, Pyrin Domain-Containing 3 Protein/analysis , Aged , Aged, 80 and over , Autopsy , Blood Vessels/pathology , COVID-19/mortality , COVID-19/pathology , COVID-19/virology , Case-Control Studies , Female , Fluorescent Antibody Technique , Humans , Male , Middle Aged
20.
Journal of Biomaterials and Tissue Engineering ; 12(4):778-787, 2022.
Article in English | Web of Science | ID: covidwho-1666523

ABSTRACT

Background and purpose: Coronavirus disease 2019 (COVID-19) was spreading all over the world. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) primarily invades and infects the lungs of humans leading to COVID-19. Mild to severe clinical symptoms such as fever, cough, and shortness of breath were existed in those patients. One of the most common changes in these patients was abnormal blood routine. However, uncertainty remains regarding the dynamic characteristics of platelet in COVID-19 patients due to limited data. Therefore, we aimed to analyze the association between dynamic characteristics of blood platelet and disease severity, and to identify new monitoring indicators to treat the COVID-19 patients.Methods:In this cohort study, 398 COVID19 patients treated in the Shenzhen Third People's hospital from December 16, 2019 to March 26, IP: 182.75.148.10 On: Thu, 20 Jan 2022 08:58:32 Copyright: American Scientific Publishers 2020 were collected and participated. All data of participants including the clinical characteristics, Delivered by Ingenta imaging and laboratory information were collected. All patients included in our study were classified as four groups (mild, common, severe, and critical types) regarding clinical symptoms and relevant severe failures based on the Diagnosis Criteria. Platelet count was examined at the baseline and every 3-5 days during hospitalization. Results: The platelet count varied with clinical classifications. The platelet count in mild type was normal without significant fluctuation. While the blood platelet count of most common and severe patients had obvious fluctuations, showing as a dynamic change that first rose and then fell to the level at admission, which was consistent with the trend of lung inflammation. Bone marrow smears further showed that bone marrow hyperplasia was normal in mild, common and severe type patients, and megakaryocytes and their platelet-producing functions were not abnormal. Conclusions: Our results suggested that the dynamic changes of platelet count might be a predictor of lung inflammation alteration for COVID-19 patients. The changes in platelet count might be a responsive pattern secondary to lung inflammation. The function of bone marrow may be slightly affected by SARS-CoV-2 infection.

SELECTION OF CITATIONS
SEARCH DETAIL