Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Ethnopharmacol ; 280: 114488, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1397458

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Traditional Chinese medicine (TCM) has a long history in the prevention and treatment of pandemics. The TCM formula Lung Cleansing and Detoxifying Decoction (LCDD), also known as Qing Fei Pai Du Decoction, has been demonstrated effective against Coronavirus Disease 2019 (COVID-19). AIM OF THE STUDY: This work aimed to elucidate the active ingredients, targets and pathway mechanism of LCDD related to suppression of inflammatory, immunity regulation and relaxation of airway smooth muscle for the treatment of COVID-19. MATERIALS AND METHODS: Mining chemical ingredients reported in LCDD, 144 compounds covering all herbs were selected and screened against inflammatory-, immunity- and respiratory-related GPCRs including GPR35, H1, CB2, B2, M3 and ß2-adrenoceptor receptor using a label-free integrative pharmacology method. Further, all active compounds were detected using liquid chromatography-tandem mass spectrometry, and an herb-compound-target network based on potency and content of compounds was constructed to elucidate the multi-target and synergistic effect. RESULTS: Thirteen compounds were identified as GPR35 agonists, including licochalcone B, isoliquiritigenin, etc. Licochalcone B, isoliquiritigenin and alisol A exhibited bradykinin receptor B2 antagonism activities. Atractyline and shogaol showed as a cannabinoid receptor CB2 agonist and a histamine receptor H1 antagonist, respectively. Tectorigenin and aristofone acted as muscarinic receptor M3 antagonists, while synephrine, ephedrine and pseudoephedrine were ß2-adrenoceptor agonists. Pathway deconvolution assays suggested activation of GPR35 triggered PI3K, MEK, JNK pathways and EGFR transactivation, and the activation of ß2-adrenoceptor mediated MEK and Ca2+. The herb-compound-target network analysis found that some compounds such as licochalcone B acted on multiple targets, and multiple components interacted with the same target such as GPR35, reflecting the synergistic mechanism of Chinese medicine. At the same time, some low-abundance compounds displayed high target activity, meaning its important role in LCDD for anti-COVID-19. CONCLUSIONS: This study elucidates the active ingredients, targets and pathways of LCDD. This is useful for elucidating multitarget synergistic action for its clinical therapeutic efficacy.


Asunto(s)
Técnicas Biosensibles/métodos , Tratamiento Farmacológico de COVID-19 , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Animales , Línea Celular Tumoral , Chalconas/farmacología , Cricetulus , Medicamentos Herbarios Chinos/análisis , Efedrina/farmacología , Células HEK293 , Humanos , Inmunidad/efectos de los fármacos , Inflamación/metabolismo , Enfermedades Pulmonares/metabolismo , Músculo Liso/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Respiración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
Life Sci ; 281: 119718, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1271709

RESUMEN

AIMS: Hypoxia, a pathophysiological condition, is profound in several cardiopulmonary diseases (CPD). Every individual's lethality to a hypoxia state differs in terms of hypoxia exposure time, dosage units and dependent on the individual's genetic makeup. Most of the proposed markers for CPD were generally aim to distinguish disease samples from normal samples. Although, as per the 2018 GOLD guidelines, clinically useful biomarkers for several cardio pulmonary disease patients in stable condition have yet to be identified. We attempt to address these key issues through the identification of Dynamic Network Biomarkers (DNB) to detect hypoxia induced early warning signals of CPD before the catastrophic deterioration. MATERIALS AND METHODS: The human microvascular endothelial tissues microarray datasets (GSE11341) of lung and cardiac expose to hypoxia (1% O2) for 3, 24 and 48 h were retrieved from the public repository. The time dependent differentially expressed genes were subjected to tissue specificity and promoter analysis to filtrate the noise levels in the networks and to dissect the tissue specific hypoxia induced genes. These filtered out genes were used to construct the dynamic segmentation networks. The hypoxia induced dynamic differentially expressed genes were validated in the lung and heart tissues of male rats. These rats were exposed to hypobaric hypoxia (simulated altitude of 25,000 or PO2 - 282 mm of Hg) progressively for 3, 24 and 48 h. KEY FINDINGS: To identify the temporal key genes regulated in hypoxia, we ranked the dominant genes based on their consolidated topological features from tissue specific networks, time dependent networks and dynamic networks. Overall topological ranking described VEGFA as a single node dynamic hub and strongly communicated with tissue specific genes to carry forward their tissue specific information. We named this type of VEGFAcentric dynamic networks as "V-DNBs". As a proof of principle, our methodology helped us to identify the V-DNBs specific for lung and cardiac tissues namely V-DNBL and V-DNBC respectively. SIGNIFICANCE: Our experimental studies identified VEGFA, SLC2A3, ADM and ENO2 as the minimum and sufficient candidates of V-DNBL. The dynamic expression patterns could be readily exploited to capture the pre disease state of hypoxia induced pulmonary vascular remodelling. Whereas in V-DNBC the minimum and sufficient candidates are VEGFA, SCL2A3, ADM, NDRG1, ENO2 and BHLHE40. The time dependent single node expansion indicates V-DNBC could also be the pre disease state pathological hallmark for hypoxia-associated cardiovascular remodelling. The network cross-talk and expression pattern between V-DNBL and V-DNBC are completely distinct. On the other hand, the great clinical advantage of V-DNBs for pre disease predictions, a set of samples during the healthy condition should suffice. Future clinical studies might further shed light on the predictive power of V-DNBs as prognostic and diagnostic biomarkers for CPD.


Asunto(s)
Cardiopatías/metabolismo , Hipoxia/metabolismo , Enfermedades Pulmonares/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Biomarcadores/metabolismo , Deterioro Clínico , Regulación de la Expresión Génica , Cardiopatías/etiología , Cardiopatías/patología , Humanos , Hipoxia/complicaciones , Hipoxia/genética , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/patología , Masculino , Ratas , Ratas Sprague-Dawley
3.
Chem Biol Interact ; 345: 109568, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1283962

RESUMEN

Nuclear factor-kappa B, involved in inflammation, host immune response, cell adhesion, growth signals, cell proliferation, cell differentiation, and apoptosis defense, is a dimeric transcription factor. Inflammation is a key component of many common respiratory disorders, including asthma, chronic obstructive pulmonary disease (COPD), bronchiectasis, and acute respiratory distress syndrome. Many basic transcription factors are found in NF-κB signaling, which is a member of the Rel protein family. Five members of this family c-REL, NF-κB2 (p100/p52), RelA (p65), NF-κB1 (p105/p50), RelB, and RelA (p65) produce 5 transcriptionally active molecules. Proinflammatory cytokines, T lymphocyte, and B lymphocyte cell mitogens, lipopolysaccharides, bacteria, viral proteins, viruses, double-stranded RNA, oxidative stress, physical exertion, various chemotherapeutics are the stimulus responsible for NF-κB activation. NF-κB act as a principal component for several common respiratory illnesses, such as asthma, lung cancer, pulmonary fibrosis, COPD as well as infectious diseases like pneumonia, tuberculosis, COVID-19. Inflammatory lung disease, especially COVID-19, can make NF-κB a key target for drug production.


Asunto(s)
Enfermedades Pulmonares/metabolismo , FN-kappa B/metabolismo , Animales , Humanos , Inflamación/complicaciones , Enfermedades Pulmonares/complicaciones , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/inmunología
4.
Elife ; 102021 03 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1196112

RESUMEN

Measures of lung function are heritable, and thus, we sought to utilise genetics to propose drug-repurposing candidates that could improve respiratory outcomes. Lung function measures were found to be genetically correlated with seven druggable biochemical traits, with further evidence of a causal relationship between increased fasting glucose and diminished lung function. Moreover, we developed polygenic scores for lung function specifically within pathways with known drug targets and investigated their relationship with pulmonary phenotypes and gene expression in independent cohorts to prioritise individuals who may benefit from particular drug-repurposing opportunities. A transcriptome-wide association study (TWAS) of lung function was then performed which identified several drug-gene interactions with predicted lung function increasing modes of action. Drugs that regulate blood glucose were uncovered through both polygenic scoring and TWAS methodologies. In summary, we provided genetic justification for a number of novel drug-repurposing opportunities that could improve lung function.


Chronic respiratory disorders like asthma affect around 600 million people worldwide. Although these illnesses are widespread, they can have several different underlying causes, making them difficult to treat. Drugs that work well on one type of respiratory disorder may be completely ineffective on another. Understanding the biological and environmental factors that cause these illnesses will allow them to be treated more effectively by tailoring therapies to each patient. Reduced lung function is a factor in respiratory disorders and it can have many genetic causes. Studying the genes of patients with reduced lung function can reveal the genes involved, some of which may already be targets of existing drugs for other illnesses. So, could a patient's genetics be used to repurpose existing drugs to treat their respiratory disorders? Reay et al. combined three methods to link genetics and biological processes to the causes of reduced lung function. The results reveal several factors that could lead to new treatments. In one example, reduced lung function showed a link to genes associated with high blood sugar. As such, treatments used in diabetes might help improve lung function in some patients. Reay et al. also developed a scoring system that could predict the efficacy of a treatment based on a patient's genetics. The study suggests that COVID-19 infection could be affected by blood sugar levels too. Chronic respiratory disorders are a critical issue worldwide and have proven difficult to treat, but these results suggest a way to identify new therapies and target them to the right patients. The findings also support a connection between lung function and blood sugar levels. This implies that perhaps existing diabetes treatments ­ including diet and lifestyle changes aimed at reducing or limiting blood sugar ­ could be repurposed to treat respiratory disorders in some patients. The next step will be to perform clinical trials to test whether these therapies are in fact effective.


Asunto(s)
Reposicionamiento de Medicamentos/métodos , Hiperglucemia/genética , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/genética , Glucemia/metabolismo , Causalidad , Bases de Datos Genéticas , Estudio de Asociación del Genoma Completo/métodos , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/fisiología , Pulmón/fisiopatología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/fisiopatología , Herencia Multifactorial , Fenotipo , Polimorfismo de Nucleótido Simple , Pruebas de Función Respiratoria/métodos , Transcriptoma
5.
Oxid Med Cell Longev ; 2021: 6646923, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1093883

RESUMEN

Inflammatory lung disease results in a high global burden of death and disability. There are no effective treatments for the most severe forms of many inflammatory lung diseases, such as chronic obstructive pulmonary disease, emphysema, corticosteroid-resistant asthma, and coronavirus disease 2019; hence, new treatment options are required. Here, we review the role of oxidative imbalance in the development of difficult-to-treat inflammatory lung diseases. The inflammation-induced overproduction of reactive oxygen species (ROS) means that endogenous antioxidants may not be sufficient to prevent oxidative damage, resulting in an oxidative imbalance in the lung. In turn, intracellular signaling events trigger the production of proinflammatory mediators that perpetuate and aggravate the inflammatory response and may lead to tissue damage. The production of high levels of ROS in inflammatory lung diseases can induce the phosphorylation of mitogen-activated protein kinases, the inactivation of phosphoinositide 3-kinase (PI3K) signaling and histone deacetylase 2, a decrease in glucocorticoid binding to its receptor, and thus resistance to glucocorticoid treatment. Hence, antioxidant treatment might be a therapeutic option for inflammatory lung diseases. Preclinical studies have shown that antioxidants (alone or combined with anti-inflammatory drugs) are effective in the treatment of inflammatory lung diseases, although the clinical evidence of efficacy is weaker. Despite the high level of evidence for the efficacy of antioxidants in the treatment of inflammatory lung diseases, the discovery and clinical investigation of safer, more efficacious compounds are now a priority.


Asunto(s)
Antioxidantes/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico , Enfermedades Pulmonares/metabolismo , Animales , Humanos , Inflamación/inmunología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Enfermedades Pulmonares/inmunología , Oxidación-Reducción/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Biochem Pharmacol ; 185: 114431, 2021 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1051487

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is the binding-site and entry-point for SARS-CoV-2 in human and highly expressed in the lung. Cigarette smoking (CS) is the leading cause of pulmonary and cardiovascular diseases. Chronic CS leads to upregulation of bronchial ACE2 inducing a high vulnerability in COVID-19 smoker patients. Interestingly, CS-induced dysregulation of pulmonary renin-angiotensin system (RAS) in part contributing into the potential pathogenesis COVID-19 pneumonia and acute respiratory distress syndrome (ARDS). Since, CS-mediated ACE2 activations is not the main pathway for increasing the risk of COVID-19, it appeared that AngII/AT1R might induce an inflammatory-burst in COVID-19 response by up-regulating cyclic nucleotide phosphodiesterase type 4 (PDE4), which hydrolyses specifically the second intracellular messenger 3', 5'-cyclic AMP (cAMP). It must be pointed out that CS might induce PDE4 up-regulation similarly to the COVID-19 inflammation, and therefore could potentiate COVID-19 inflammation opening the potential therapeutic effects of PDE4 inhibitor in both COVID-19-inflammation and CS.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Fumar Cigarrillos/tratamiento farmacológico , Enfermedades Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , COVID-19/epidemiología , COVID-19/metabolismo , Fumar Cigarrillos/epidemiología , Fumar Cigarrillos/metabolismo , Humanos , Pulmón/fisiología , Enfermedades Pulmonares/epidemiología , Enfermedades Pulmonares/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/epidemiología , Síndrome de Dificultad Respiratoria/metabolismo
8.
Curr Drug Targets ; 22(13): 1477-1484, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1048853

RESUMEN

BACKGROUND: Activation of Poly (ADP-ribose) polymerase 1 (PARP1), a post-translational modifying enzyme, has been shown to be involved with several inflammatory and viral diseases. OBJECTIVES: The goal of this review is to highlight the mechanisms underlying PARP1 activation during viral or infectious pathogenesis and to assess potential possibilities of using PARP1 inhibitors as a therapeutic countering of SARS-CoV-2 virus. METHODS: An extensive bibliographic search was done using Pubmed, Mendeley and google scholar with key words. Pre-prints are reported with potential caveats and studies without experimental data were excluded. RESULTS: Covid-19, a global pandemic; is associated with systemic surge of inflammatory cytokines resulting in severe inflammation of the lung, heart dysfunction, ischemia, and stroke. PARP1 regulates expression of NFkB and downstream cytokine production and its inhibition is known to attenuate the expression of inflammatory cytokines. PARP1 and other PARP family members regulate viral infection, replication, and virulence. The literature clearly suggests that PARP1 plays an important role in host-pathogen interactions and pathogenesis, with pre-clinical and in vitro studies supporting the idea that PARP1 inhibition may negatively affect viability of several viruses including the replication of the SARS-CoV and SARS-CoV-2 virus. CONCLUSION: The current review discusses mechanisms of PARP1 activation during viral infection, inflammatory diseases, cytokine expression and possibility of PARP1 in regulating cytokine storm and hyper-inflammation seen with Covid-19. Additionally, in vitro studies showing the negative regulation of SARS-CoV-2 virus replication by PARP inhibitors indicates a potential therapeutic role of PARP inhibitors for Covid-19 or its variants.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/virología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/metabolismo , SARS-CoV-2/enzimología , Animales , Enfermedades Cardiovasculares/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Enfermedades Pulmonares/metabolismo , Poli(ADP-Ribosa) Polimerasas/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo
9.
Thorax ; 76(1): 92-99, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-978824

RESUMEN

The lungs are exposed to a range of environmental toxins (including cigarette smoke, air pollution, asbestos) and pathogens (bacterial, viral and fungal), and most respiratory diseases are associated with local or systemic hypoxia. All of these adverse factors can trigger endoplasmic reticulum (ER) stress. The ER is a key intracellular site for synthesis of secretory and membrane proteins, regulating their folding, assembly into complexes, transport and degradation. Accumulation of misfolded proteins within the lumen results in ER stress, which activates the unfolded protein response (UPR). Effectors of the UPR temporarily reduce protein synthesis, while enhancing degradation of misfolded proteins and increasing the folding capacity of the ER. If successful, homeostasis is restored and protein synthesis resumes, but if ER stress persists, cell death pathways are activated. ER stress and the resulting UPR occur in a range of pulmonary insults and the outcome plays an important role in many respiratory diseases. The UPR is triggered in the airway of patients with several respiratory diseases and in corresponding experimental models. ER stress has been implicated in the initiation and progression of pulmonary fibrosis, and evidence is accumulating suggesting that ER stress occurs in obstructive lung diseases (particularly in asthma), in pulmonary infections (some viral infections and in the setting of the cystic fibrosis airway) and in lung cancer. While a number of small molecule inhibitors have been used to interrogate the role of the UPR in disease models, many of these tools have complex and off-target effects, hence additional evidence (eg, from genetic manipulation) may be required to support conclusions based on the impact of such pharmacological agents. Aberrant activation of the UPR may be linked to disease pathogenesis and progression, but at present, our understanding of the context-specific and disease-specific mechanisms linking these processes is incomplete. Despite this, the ability of the UPR to defend against ER stress and influence a range of respiratory diseases is becoming increasingly evident, and the UPR is therefore attracting attention as a prospective target for therapeutic intervention strategies.


Asunto(s)
Estrés del Retículo Endoplásmico , Enfermedades Pulmonares/metabolismo , Proteínas de la Membrana/fisiología , Humanos , Transducción de Señal
10.
Respir Res ; 21(1): 276, 2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: covidwho-883579

RESUMEN

BACKGROUND: Severe coronavirus disease 2019 (COVID-19) is frequently associated with hyperinflammation and hyperferritinemia. The latter is related to increased mortality in COVID-19. Still, it is not clear if iron dysmetabolism is mechanistically linked to COVID-19 pathobiology. METHODS: We herein present data from the ongoing prospective, multicentre, observational CovILD cohort study (ClinicalTrials.gov number, NCT04416100), which systematically follows up patients after COVID-19. 109 participants were evaluated 60 days after onset of first COVID-19 symptoms including clinical examination, chest computed tomography and laboratory testing. RESULTS: We investigated subjects with mild to critical COVID-19, of which the majority received hospital treatment. 60 days after disease onset, 30% of subjects still presented with iron deficiency and 9% had anemia, mostly categorized as anemia of inflammation. Anemic patients had increased levels of inflammation markers such as interleukin-6 and C-reactive protein and survived a more severe course of COVID-19. Hyperferritinemia was still present in 38% of all individuals and was more frequent in subjects with preceding severe or critical COVID-19. Analysis of the mRNA expression of peripheral blood mononuclear cells demonstrated a correlation of increased ferritin and cytokine mRNA expression in these patients. Finally, persisting hyperferritinemia was significantly associated with severe lung pathologies in computed tomography scans and a decreased performance status as compared to patients without hyperferritinemia. DISCUSSION: Alterations of iron homeostasis can persist for at least two months after the onset of COVID-19 and are closely associated with non-resolving lung pathologies and impaired physical performance. Determination of serum iron parameters may thus be a easy to access measure to monitor the resolution of COVID-19. TRIAL REGISTRATION: ClinicalTrials.gov number: NCT04416100.


Asunto(s)
Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/metabolismo , Homeostasis , Hierro/metabolismo , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/metabolismo , Neumonía Viral/complicaciones , Neumonía Viral/metabolismo , Adulto , Anciano , Anemia/etiología , Proteína C-Reactiva/análisis , COVID-19 , Estudios de Cohortes , Infecciones por Coronavirus/fisiopatología , Femenino , Ferritinas/sangre , Estudios de Seguimiento , Humanos , Inflamación/etiología , Inflamación/metabolismo , Interleucina-6/sangre , Enfermedades Pulmonares/fisiopatología , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Pandemias , Neumonía Viral/fisiopatología , Estudios Prospectivos , Tomografía Computarizada por Rayos X
11.
Nat Commun ; 11(1): 5165, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: covidwho-872694

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) has been identified as the host entry receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the COVID-19 pandemic. ACE2 is a regulatory enzyme of the renin-angiotensin system and has protective functions in many cardiovascular, pulmonary and metabolic diseases. This review summarizes available murine models with systemic or organ-specific deletion of ACE2, or with overexpression of murine or human ACE2. The purpose of this review is to provide researchers with the genetic tools available for further understanding of ACE2 biology and for the investigation of ACE2 in the pathogenesis and treatment of COVID-19.


Asunto(s)
Enfermedades Cardiovasculares/patología , Modelos Animales de Enfermedad , Enfermedades Pulmonares/patología , Peptidil-Dipeptidasa A/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/fisiología , COVID-19 , Enfermedades Cardiovasculares/metabolismo , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Humanos , Enfermedades Pulmonares/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Ratones , Ratones Mutantes , Pandemias , Peptidil-Dipeptidasa A/genética , Neumonía Viral/metabolismo , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2
12.
Eur Respir Rev ; 29(157)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: covidwho-810448

RESUMEN

The respiratory tract and its resident immune cells face daily exposure to stress, both from without and from within. Inhaled pathogens, including severe acute respiratory syndrome coronavirus 2, and toxins from pollution trigger a cellular defence system that reduces protein synthesis to minimise viral replication or the accumulation of misfolded proteins. Simultaneously, a gene expression programme enhances antioxidant and protein folding machineries in the lung. Four kinases (PERK, PKR, GCN2 and HRI) sense a diverse range of stresses to trigger this "integrated stress response". Here we review recent advances identifying the integrated stress response as a critical pathway in the pathogenesis of pulmonary diseases, including pneumonias, thoracic malignancy, pulmonary fibrosis and pulmonary hypertension. Understanding the integrated stress response provides novel targets for the development of therapies.


Asunto(s)
Inflamación/metabolismo , Enfermedades Pulmonares/metabolismo , Estrés Oxidativo/fisiología , Biomarcadores/metabolismo , Humanos
13.
Kidney Blood Press Res ; 45(5): 661-670, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-781259

RESUMEN

BACKGROUND: The outbreak of severe acute respiratory syndrome ß-coronavirus 2 (SARS-CoV-2) has the potential to become a long-lasting global health crisis. The number of people infected with the novel coronavirus has surpassed 22 million globally, resulting in over 700,000 deaths with more than 15 million people having recovered (https://covid19.who.int). Enormous efforts are underway for rapid vaccine and treatment developments. Amongst the many ways of tackling the novel coronavirus disease 2019 (COVID-19) pandemic, extracellular vesicles (EVs) are emerging. SUMMARY: EVs are lipid bilayer-enclosed structures secreted from all types of cells, including those lining the respiratory tract. They have established roles in lung immunity and are involved in the pathogenesis of various lung diseases, including viral infection. In this review, we point out the roles and possible contribution of EVs in viral infections, as well as ongoing EV-based approaches for the treatment of COVID-19, including clinical trials. Key Messages: EVs share structural similarities to viruses and recent findings demonstrate that viruses exploit EVs for cellular exit and EVs exploit viral entry mechanisms for cargo delivery. Moreover, EV-virus interplay could be exploited for future antiviral drug and vaccine development. EV-based therapies, especially the mesenchymal stem cell-derived EVs, are being intensively studied for the treatment of COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/terapia , Vesículas Extracelulares/virología , Enfermedades Pulmonares/terapia , Neumonía Viral/terapia , Antivirales/administración & dosificación , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/metabolismo , Vesículas Extracelulares/metabolismo , Terapia Genética/tendencias , Humanos , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/virología , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/metabolismo , SARS-CoV-2 , Esparcimiento de Virus/efectos de los fármacos , Esparcimiento de Virus/fisiología
14.
Adv Exp Med Biol ; 1265: 57-70, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-712830

RESUMEN

Lung diseases affect millions of individuals all over the world. Various environmental factors, such as toxins, chemical pollutants, detergents, viruses, bacteria, microbial dysbiosis, and allergens, contribute to the development of respiratory disorders. Exposure to these factors activates stress responses in host cells and disrupt lung homeostasis, therefore leading to dysfunctional epithelial barriers. Despite significant advances in therapeutic treatments for lung diseases in the last two decades, novel interventional targets are imperative, considering the side effects and limited efficacy in patients treated with currently available drugs. Nutrients, such as amino acids (e.g., arginine, glutamine, glycine, proline, taurine, and tryptophan), peptides, and bioactive molecules, have attracted more and more attention due to their abilities to reduce oxidative stress, inhibit apoptosis, and regulate immune responses, thereby improving epithelial barriers. In this review, we summarize recent advances in amino acid metabolism in the lungs, as well as multifaceted functions of amino acids in attenuating inflammatory lung diseases based on data from studies with both human patients and animal models. The underlying mechanisms for the effects of physiological amino acids are likely complex and involve cell signaling, gene expression, and anti-oxidative reactions. The beneficial effects of amino acids are expected to improve the respiratory health and well-being of humans and other animals. Because viruses (e.g., coronavirus) and environmental pollutants (e.g., PM2.5 particles) induce severe damage to the lungs, it is important to determine whether dietary supplementation or intravenous administration of individual functional amino acids (e.g., arginine-HCl, citrulline, N-acetylcysteine, glutamine, glycine, proline and tryptophan) or their combinations to affected subjects may alleviate injury and dysfunction in this vital organ.


Asunto(s)
Aminoácidos/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Animales , Humanos , Enfermedades Pulmonares/fisiopatología
15.
Ann Clin Lab Sci ; 50(3): 308-313, 2020 May.
Artículo en Inglés | MEDLINE | ID: covidwho-614689

RESUMEN

OBJECTIVE: The COVID-19 pandemic has challenged the world economically and medically. Understanding and defining the biology of this specific coronavirus infection may lead to targeted therapies to lessen its virulence and expand the host resistance. This study's objective was to apply morphoproteomics to pulmonary lung sections from a forensic autopsy of an untreated COVID-19 victim, so that we may better define its biology from the perspective of its interaction with the host and provide options for therapeutic targets. DESIGN: Morphoproteomic analysis from a case study of this COVID-19 pulmonary infection included immunohistochemical probes to detect phosphorylated p-STAT3 (Tyr 705), as part of the interleukin (IL)-6 pathway; cyclooxygenase (COX)-2, CD8+ cytotoxic lymphocytes, Programmed Death (PD)-1 receptor+ lymphoid cells, CD56+ NK lymphoid cells, CD163+ (M2 polarized monocytes/macrophages), and programmed death-ligand 1 (PD-L1) expression as part of the host response to interaction with the COVID-19 virus. RESULTS: Representative sections of the COVID-19 victim's lung showed: nuclear expression of p-STAT3 (Tyr 705) in many of the alveolar pneumocytes and in occasional endothelial cells; COX-2 expression in the alveolar pneumocytes; a relative paucity of CD8+ cytotoxic lymphocytes; absence of CD56+ NK lymphoid cells; abundance of intra-alveolar and alveolar interstitial CD163+ macrophages/monocytes; PD-L1 expression on occasional macrophages, focally on collections of alveolar pneumocytes, and on cells in the alveolar interstitium; and rare PD-1+ lymphocytes in similar regions as CD8+ lymphocytes. CONCLUSION: Morphoproteomics and microanatomical features coincide with the etiopathogenic features of pulmonary coronavirus infection and the host response. This suggests that a targeted therapy could address the biology of COVID-19 pneumonia, enhance the host immune response and prevent its progression to a life-threatening, ventilator-dependent clinical situation.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Biomarcadores/metabolismo , Infecciones por Coronavirus/complicaciones , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Neumonía Viral/complicaciones , Proteoma/análisis , Biomarcadores/análisis , COVID-19 , Infecciones por Coronavirus/virología , Resultado Fatal , Humanos , Enfermedades Pulmonares/etiología , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/virología , Proteoma/metabolismo , SARS-CoV-2
16.
J Exp Med ; 217(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: covidwho-607919

RESUMEN

The renin-angiotensin system (RAS) has long been appreciated as a major regulator of blood pressure, but has more recently been recognized as a mechanism for modulating inflammation as well. While there has been concern in COVID-19 patients over the use of drugs that target this system, the RAS has not been explored fully as a druggable target. The abbreviated description of the RAS suggests that its dysregulation may be at the center of COVID-19.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Enfermedades Pulmonares/fisiopatología , Pulmón/virología , Neumonía Viral/fisiopatología , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Trastornos de la Coagulación Sanguínea/virología , COVID-19 , Infecciones por Coronavirus/etiología , Infecciones por Coronavirus/metabolismo , Citocinas/metabolismo , Humanos , Hipertensión/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/virología , Obesidad/fisiopatología , Pandemias , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/etiología , Neumonía Viral/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Índice de Severidad de la Enfermedad
17.
Int J Mol Sci ; 21(9)2020 Apr 28.
Artículo en Inglés | MEDLINE | ID: covidwho-133432

RESUMEN

BACKGROUND: On the 31 December 2019, the World Health Organization (WHO) was informed of a cluster of cases of pneumonia of unknown origin detected in Wuhan City, Hubei Province, China. The infection spread first in China and then in the rest of the world, and on the 11th of March, the WHO declared that COVID-19 was a pandemic. Taking into consideration the mortality rate of COVID-19, about 5-7%, and the percentage of positive patients admitted to intensive care units being 9-11%, it should be mandatory to consider and take all necessary measures to contain the COVID-19 infection. Moreover, given the recent evidence in different hospitals suggesting IL-6 and TNF-α inhibitor drugs as a possible therapy for COVID-19, we aimed to highlight that a dietary intervention could be useful to prevent the infection and/or to ameliorate the outcomes during therapy. Considering that the COVID-19 infection can generate a mild or highly acute respiratory syndrome with a consequent release of pro-inflammatory cytokines, including IL-6 and TNF-α, a dietary regimen modification in order to improve the levels of adiponectin could be very useful both to prevent the infection and to take care of patients, improving their outcomes.


Asunto(s)
Antioxidantes/administración & dosificación , Betacoronavirus , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/terapia , Dieta , Suplementos Dietéticos , Neumonía Viral/inmunología , Neumonía Viral/terapia , Adiponectina/metabolismo , Ácido Ascórbico/administración & dosificación , COVID-19 , Infecciones por Coronavirus/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Ácidos Grasos Omega-3/metabolismo , Flavonoides/administración & dosificación , Humanos , Interleucina-6/inmunología , Interleucina-6/metabolismo , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/terapia , Pandemias , Neumonía Viral/metabolismo , SARS-CoV-2 , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Exp Med ; 217(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: covidwho-72158

RESUMEN

Coronavirus disease 2019 (COVID-19) is a novel, viral-induced respiratory disease that in ∼10-15% of patients progresses to acute respiratory distress syndrome (ARDS) triggered by a cytokine storm. In this Perspective, autopsy results and literature are presented supporting the hypothesis that a little known yet powerful function of neutrophils-the ability to form neutrophil extracellular traps (NETs)-may contribute to organ damage and mortality in COVID-19. We show lung infiltration of neutrophils in an autopsy specimen from a patient who succumbed to COVID-19. We discuss prior reports linking aberrant NET formation to pulmonary diseases, thrombosis, mucous secretions in the airways, and cytokine production. If our hypothesis is correct, targeting NETs directly and/or indirectly with existing drugs may reduce the clinical severity of COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/patología , Trampas Extracelulares , Enfermedades Pulmonares , Neutrófilos/patología , Neumonía Viral/patología , COVID-19 , Infecciones por Coronavirus/complicaciones , Citocinas/metabolismo , Humanos , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Pandemias , Neumonía Viral/complicaciones , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA