Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Hazard Mater ; 456: 131654, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: covidwho-2320886

RESUMEN

Functional face masks that can effectively remove particulate matter and pathogens are critical to addressing the urgent health needs arising from industrial air pollution and the COVID-19 pandemic. However, most commercial masks are manufactured by tedious and complicated network-forming procedures (e.g., meltblowing and electrospinning). In addition, the materials used (e.g., polypropylene) have significant limitations such as a lack of pathogen inactivation and degradability, which can cause secondary infection and serious environmental concerns if discarded. Here, we present a facile and straightforward method for creating biodegradable and self-disinfecting masks based on collagen fiber networks. These masks not only provide superior protection against a wide range of hazardous substances in polluted air, but also address environmental concerns associated with waste disposal. Importantly, collagen fiber networks with naturally existing hierarchical microporous structures can be easily modified by tannic acid to improve its mechanical characteristics and enable the in situ production of silver nanoparticles. The resulting masks exhibit excellent antibacterial (>99.99%, 15 min) and antiviral (>99.999%, 15 min) capabilities, as well as high PM2.5 removal efficiency (>99.9%, 30 s). We further demonstrate the integration of the mask into a wireless platform for respiratory monitoring. Therefore, the smart mask has enormous promise for combating air pollution and contagious viruses, managing personal health, and alleviating waste issues caused by commercial masks.


Asunto(s)
COVID-19 , Nanopartículas del Metal , Humanos , Antivirales , Pandemias/prevención & control , COVID-19/prevención & control , Plata , Polvo , Antibacterianos/farmacología , Colágeno
2.
Int J Mol Sci ; 24(9)2023 May 06.
Artículo en Inglés | MEDLINE | ID: covidwho-2320574

RESUMEN

Extracellular collagen remodeling is one of the central mechanisms responsible for the structural and compositional coherence of myocardium in patients undergoing myocardial infarction (MI). Activated primary cardiac fibroblasts following myocardial infarction are extensively investigated to establish anti-fibrotic therapies to improve left ventricular remodeling. To systematically assess vitamin C functions as a potential modulator involved in collagen fibrillogenesis in an in vitro model mimicking heart tissue healing after MI. Mouse primary cardiac fibroblasts were isolated from wild-type C57BL/6 mice and cultured under normal and profibrotic (hypoxic + transforming growth factor beta 1) conditions on freshly prepared coatings mimicking extracellular matrix (ECM) remodeling during healing after an MI. At 10 µg/mL, vitamin C reprogramed the respiratory mitochondrial metabolism, which is effectively associated with a more increased accumulation of intracellular reactive oxygen species (iROS) than the number of those generated by mitochondrial reactive oxygen species (mROS). The mRNA/protein expression of subtypes I, III collagen, and fibroblasts differentiations markers were upregulated over time, particularly in the presence of vitamin C. The collagen substrate potentiated the modulator role of vitamin C in reinforcing the structure of types I and III collagen synthesis by reducing collagen V expression in a timely manner, which is important in the initiation of fibrillogenesis. Altogether, our study evidenced the synergistic function of vitamin C at an optimum dose on maintaining the equilibrium functionality of radical scavenger and gene transcription, which are important in the initial phases after healing after an MI, while modulating the synthesis of de novo collagen fibrils, which is important in the final stage of tissue healing.


Asunto(s)
Ácido Ascórbico , Infarto del Miocardio , Ratones , Animales , Ácido Ascórbico/farmacología , Ácido Ascórbico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Vitaminas/metabolismo , Remodelación Ventricular/fisiología
3.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: covidwho-2249413

RESUMEN

Myeloperoxidase is an enzyme released by neutrophils when neutrophil extracellular traps (NETs) are formed. Besides myeloperoxidase activity against pathogens, it was also linked to many diseases, including inflammatory and fibrotic ones. Endometrosis is a fibrotic disease of the mare endometrium, with a large impact on their fertility, where myeloperoxidase was shown to induce fibrosis. Noscapine is an alkaloid with a low toxicity, that has been studied as an anti-cancer drug, and more recently as an anti-fibrotic molecule. This work aims to evaluate noscapine inhibition of collagen type 1 (COL1) induced by myeloperoxidase in equine endometrial explants from follicular and mid-luteal phases, at 24 and 48 h of treatment. The transcription of collagen type 1 alpha 2 chain (COL1A2), and COL1 protein relative abundance were evaluated by qPCR and Western blot, respectively. The treatment with myeloperoxidase increased COL1A2 mRNA transcription and COL1 protein, whereas noscapine was able to reduce this effect with respect to COL1A2 mRNA transcription, in a time/estrous cycle phase-dependent manner (in explants from the follicular phase, at 24 h of treatment). Our study indicates that noscapine is a promising drug to be considered as an anti-fibrotic molecule to prevent endometrosis development, making noscapine a strong candidate to be applied in future endometrosis therapies.


Asunto(s)
Fibrosis , Noscapina , Peroxidasa , Animales , Femenino , Colágeno/metabolismo , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Fibrosis/veterinaria , Caballos/metabolismo , Noscapina/farmacología , Noscapina/uso terapéutico , Peroxidasa/antagonistas & inhibidores , Peroxidasa/metabolismo , ARN Mensajero/metabolismo
4.
Front Immunol ; 14: 1130288, 2023.
Artículo en Inglés | MEDLINE | ID: covidwho-2259138

RESUMEN

Introduction: Thromboinflammatory complications are well described sequalae of Coronavirus Disease 2019 (COVID-19), and there is evidence of both hyperreactive platelet and inflammatory neutrophil biology that contributes to the thromoinflammatory milieu. It has been demonstrated in other thromboinflammatory diseases that the circulating environment may affect cellular behavior, but what role this environment exerts on platelets and neutrophils in COVID-19 remains unknown. We tested the hypotheses that 1) plasma from COVID-19 patients can induce a prothrombotic platelet functional phenotype, and 2) contents released from platelets (platelet releasate) from COVID-19 patients can induce a proinflammatory neutrophil phenotype. Methods: We treated platelets with COVID-19 patient and disease control plasma, and measured their aggregation response to collagen and adhesion in a microfluidic parallel plate flow chamber coated with collagen and thromboplastin. We exposed healthy neutrophils to platelet releasate from COVID-19 patients and disease controls and measured neutrophil extracellular trap formation and performed RNA sequencing. Results: We found that COVID-19 patient plasma promoted auto-aggregation, thereby reducing response to further stimulation ex-vivo. Neither disease condition increased the number of platelets adhered to a collagen and thromboplastin coated parallel plate flow chamber, but both markedly reduced platelet size. COVID-19 patient platelet releasate increased myeloperoxidasedeoxyribonucleic acid complexes and induced changes to neutrophil gene expression. Discussion: Together these results suggest aspects of the soluble environment circulating platelets, and that the contents released from those neutrophil behavior independent of direct cellular contact.


Asunto(s)
Plaquetas , COVID-19 , Humanos , Plaquetas/metabolismo , Neutrófilos/metabolismo , COVID-19/metabolismo , Tromboplastina/metabolismo , Colágeno/metabolismo
5.
J Am Heart Assoc ; 12(4): e027990, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: covidwho-2244399

RESUMEN

Background Cardiac fibrosis complicates SARS-CoV-2 infections and has been linked to arrhythmic complications in survivors. Accordingly, we sought evidence of increased HSP47 (heat shock protein 47), a stress-inducible chaperone protein that regulates biosynthesis and secretion of procollagen in heart tissue, with the goal of elucidating molecular mechanisms underlying cardiac fibrosis in subjects with this viral infection. Methods and Results Using human autopsy tissue, immunofluorescence, and immunohistochemistry, we quantified Hsp47+ cells and collagen α 1(l) in hearts from people with SARS-CoV-2 infections. Because macrophages are also linked to inflammation, we measured CD163+ cells in the same tissues. We observed irregular groups of spindle-shaped HSP47+ and CD163+ cells as well as increased collagen α 1(I) deposition, each proximate to one another in "hot spots" of ≈40% of hearts after SARS-CoV-2 infection (HSP47+ P<0.05 versus nonfibrotics and P<0.001 versus controls). Because HSP47+ cells are consistent with myofibroblasts, subjects with hot spots are termed "profibrotic." The remaining 60% of subjects dying with COVID-19 without hot spots are referred to as "nonfibrotic." No control subject exhibited hot spots. Conclusions Colocalization of myofibroblasts, M2(CD163+) macrophages, and collagen α 1(l) may be the first evidence of a COVID-19-related "profibrotic phenotype" in human hearts in situ. The potential public health and diagnostic implications of these observations require follow-up to further define mechanisms of viral-mediated cardiac fibrosis.


Asunto(s)
COVID-19 , Miofibroblastos , Humanos , Miofibroblastos/metabolismo , SARS-CoV-2 , Colágeno/metabolismo , Proteínas de Choque Térmico/metabolismo , Colágeno Tipo I/metabolismo , Fenotipo , Macrófagos/metabolismo , Fibrosis
6.
J Thromb Haemost ; 21(1): 94-100, 2023 01.
Artículo en Inglés | MEDLINE | ID: covidwho-2210985

RESUMEN

BACKGROUND: Post-COVID syndrome (PCS) affects millions of people worldwide, causing a multitude of symptoms and impairing quality of life months or even years after acute COVID-19. A prothrombotic state has been suggested; however, underlying mechanisms remain to be elucidated. OBJECTIVES: To investigate thrombogenicity in PCS using a microfluidic assay, linking microthrombi, thrombin generation, and the von Willebrand factor (VWF):a Disintegrin and Metalloproteinase with a Thrombospondin Type 1 motif, member 13 (ADAMTS13) axis. METHODS: Citrated blood was perfused through microfluidic channels coated with collagen or an antibody against the VWF A3 domain, and thrombogenicity was monitored in real time. Thrombin generation assays were performed and α(2)-antiplasmin, VWF, and ADAMTS13 activity levels were also measured. RESULTS: We investigated thrombogenicity in a cohort of 21 patients with PCS with a median time following symptoms onset of 23 months using a dynamic microfluidic assay. Our data show a significant increase in platelet binding on both collagen and anti-VWF A3 in patients with PCS compared with that in controls, which positively correlated with VWF antigen (Ag) levels, the VWF(Ag):ADAMTS13 ratio (on anti-VWF A3), and inversely correlated with ADAMTS13 activity (on collagen). Thrombi forming on collagen presented different geometries in patients with PCS vs controls, with significantly increased thrombi area mainly attributable to thrombi length in the patient group. Thrombi length positively correlated with VWF(Ag):ADAMTS13 ratio and thrombin generation assay results, which were increased in 55.5% of patients. α(2)-Antiplasmin levels were normal in 89.5% of patients. CONCLUSION: Together, these data present a dynamic assay to investigate the prothrombotic state in PCS, which may help unravel the mechanisms involved and/or establish new therapeutic strategies for this condition.


Asunto(s)
Antifibrinolíticos , COVID-19 , Trombosis , Humanos , Trombina , Calidad de Vida , Proteínas ADAM/metabolismo , COVID-19/complicaciones , Factor de von Willebrand/metabolismo , Trombosis/etiología , Colágeno , Proteína ADAMTS13
7.
Molecules ; 28(2)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: covidwho-2200545

RESUMEN

Pulmonary fibrosis (PF) is one of the sequelae of Corona Virus Disease 2019 (COVID-19), and currently, lung transplantation is the only viable treatment option. Hence, other effective treatments are urgently required. We investigated the therapeutic effects of an approved botanical drug, cepharanthine (CEP), in a cell culture model of transforming growth factor-ß1 (TGF-ß1) and bleomycin (BLM)-induced pulmonary fibrosis rat models both in vitro and in vivo. In this study, CEP and pirfenidone (PFD) suppressed BLM-induced lung tissue inflammation, proliferation of blue collagen fibers, and damage to lung structures in vivo. Furthermore, we also found increased collagen deposition marked by α-smooth muscle actin (α-SMA) and Collagen Type I Alpha 1 (COL1A1), which was significantly alleviated by the addition of PFD and CEP. Moreover, we elucidated the underlying mechanism of CEP against PF in vitro. Various assays confirmed that CEP reduced the viability and migration and promoted apoptosis of myofibroblasts. The expression levels of myofibroblast markers, including COL1A1, vimentin, α-SMA, and Matrix Metallopeptidase 2 (MMP2), were also suppressed by CEP. Simultaneously, CEP significantly suppressed the elevated Phospho-NF-κB p65 (p-p65)/NF-κB p65 (p65) ratio, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) levels, and elevated inhibitor of NF-κB Alpha (IκBα) degradation and reversed the progression of PF. Hence, our study demonstrated that CEP prevented myofibroblast activation and treated BLM-induced pulmonary fibrosis in a dose-dependent manner by regulating nuclear factor kappa-B (NF-κB)/ NLRP3 signaling, thereby suggesting that CEP has potential clinical application in pulmonary fibrosis in the future.


Asunto(s)
COVID-19 , Fibrosis Pulmonar , Animales , Ratas , Bleomicina , Colágeno/metabolismo , COVID-19/metabolismo , Fibroblastos/metabolismo , Inflamación/metabolismo , Pulmón , Miofibroblastos/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
8.
Cells ; 12(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: covidwho-2166271

RESUMEN

Critically ill COVID-19 patients suffer from thromboembolic as well as bleeding events. Endothelial dysfunction, spiking of von Willebrand factor (vWF), and excessive cytokine signaling result in coagulopathy associated with substantial activation of plasmatic clotting factors. Thrombocytopenia secondary to extensive platelet activation is a frequent finding, but abnormal platelet dysfunction may also exist in patients with normal platelet counts. In this study, we performed analyses of platelet function and of von Willebrand factor in critically ill COVID-19 patients (n = 13). Platelet aggregometry was performed using ADP, collagen, epinephrin, and ristocetin. VWF and fibrinogen binding of platelets and CD62 and CD63 expression after thrombin stimulation were analyzed via flow cytometry. In addition, VWF antigen (VWF:Ag), collagen binding capacity (VWF:CB), and multimer analysis were performed next to routine coagulation parameters. All patients exhibited reduced platelet aggregation and decreased CD62 and CD63 expression. VWF binding of platelets was reduced in 12/13 patients. VWF:CB/VWF:Ag ratios were pathologically decreased in 2/13 patients and elevated in 2/13 patients. Critically ill COVID-19 patients exhibit platelet secretion defects independent of thrombocytopenia. Platelet exhaustion and VWF dysfunction may result in impaired primary hemostasis and should be considered when treating coagulopathy in these patients.


Asunto(s)
COVID-19 , Trombocitopenia , Humanos , Factor de von Willebrand/metabolismo , SARS-CoV-2/metabolismo , Enfermedad Crítica , Agregación Plaquetaria , COVID-19/complicaciones , Hemostasis , Trombocitopenia/complicaciones , Colágeno/metabolismo
9.
Curr Top Microbiol Immunol ; 436: 437-466, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2075205

RESUMEN

A number of different experimental models using both non-selective and selective PI3K inhibitors have shown that many pathogenic steps of respiratory disorders, such as bronchial asthma, Chronic Obstructive Pulmonary Disease (COPD), Idiopathic Pulmonary Fibrosis (IPF), Acute Respiratory Distress Syndrome (ARDS) and Lung Cancer (LC) are, at least in part, regulated by the PI3K signaling pathway, suggesting that the inhibition of PI3K could represent an ideal therapeutic target for the treatment of respiratory diseases. This chapter summarizes the current state of the therapeutic strategies aimed to exploit the inhibition of PI3K in this context. In animal models of asthma, selective δ and γ inhibitors have shown to be effective, and when administered by inhalation, reasonably safe. Nevertheless, very few clinical trials have been performed so far. The efficacy of current traditional therapies for allergic bronchial asthma has likely diminished the need for new alternative treatments. Surprisingly, in COPD, where instead there is an urgent need for new and more effective therapeutic approaches, the number of clinical studies is still low and not capable yet, with the exception for an acceptable safety profile, to show a significant improvement of clinical outcomes. In IPF, a disease with a disappointing prognosis, PI3K inhibitors have been bound to a FAP ligand with the aim to selectively target myofibroblasts, showing to significantly reduce collagen production and the development of lung fibrosis in an animal model of lung fibrosis. Due to its role in cell activation and cell replication, the PI3K pathway is obviously largely involved in lung cancer. Several studies, currently ongoing, are testing the effect of PI3K inhibitors mainly in NSCLC. Some evidence in the treatment of cancer patients suggests the possibility that PI3K inhibitors may enhance the response to conventional treatment. The involvement of PI3Kδ in the modulation of airway neutrophil recruitment and bronchial epithelial functional alterations also suggest a potential role in the treatment of ARDS, but at the current state the ongoing trials are aimed to the treatment of ARDS in COVID-19 patients. In general, few clinical trials investigating PI3K inhibitors in respiratory disorders have been performed so far. This relatively new approach of treatment is just at its beginning and certainly needs further efforts and additional studies.


Asunto(s)
Asma , COVID-19 , Fibrosis Pulmonar Idiopática , Neoplasias Pulmonares , Enfermedad Pulmonar Obstructiva Crónica , Síndrome de Dificultad Respiratoria , Animales , Asma/tratamiento farmacológico , Colágeno/uso terapéutico , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Ligandos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
10.
Mol Med Rep ; 26(5)2022 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-2055488

RESUMEN

COVID­19 patients with severe infection have been observed to have elevated auto­antibodies (AAs) against angiotensin II receptor type 1 (AT1R) and endothelin (ET) 1 receptor type A (ETAR), compared with healthy controls and patients with favorable (mild) infection. AT1R and ETAR are G protein­coupled receptors, located on vascular smooth muscle cells, fibroblasts, immune and endothelial cells, and are activated by angiotensin II (Ang II) and ET1 respectively. AAs that are specific for these receptors have a functional role similar to the natural ligands, but with a more prolonged vasoconstrictive effect. They also induce the production of fibroblast collagen, the release of reactive oxygen species and the secretion of proinflammatory cytokines (including IL­6, IL­8 and TNF­α) by immune cells. Despite the presence of AAs in severe COVID­19 infected patients, their contribution and implication in the severity of the disease is still not well understood and further studies are warranted. The present review described the major vascular homeostasis systems [ET and renin­angiotensin­aldosterone system (RAAS)], the vital regulative role of nitric oxide, the AAs, and finally the administration of angiotensin II receptor blockers (ARBs), so as to provide more insight into the interplay that exists among these components and their contribution to the severity, prognosis and possible treatment of COVID­19.


Asunto(s)
COVID-19 , Enfermedades Vasculares , Angiotensina II , Antagonistas de Receptores de Angiotensina , Inhibidores de la Enzima Convertidora de Angiotensina , Colágeno , Células Endoteliales , Endotelinas , Humanos , Interleucina-6 , Interleucina-8 , Óxido Nítrico , Especies Reactivas de Oxígeno , Receptor de Angiotensina Tipo 1 , Receptor de Endotelina A , Receptores de Angiotensina , Factor de Necrosis Tumoral alfa
11.
Arch Oral Biol ; 144: 105553, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-2048915

RESUMEN

OBJECTIVE: This systematic review examines the effect of age on changes to coronal dentin properties. DESIGN: Pubmed, Cinhal, Scopus, Web of Science and the Cochrane Database were searched for publications up to 31 December 2021. All studies were uploaded and reviewed using Covidence software. At different stages of the review, study selection and the extraction of data were completed by six independent reviewers based on the eligibility criteria. The quality of the articles was judged based on JBI Critical Appraisal Checklist for quasi-experimental studies. RESULTS: Twelve studies satisfied the eligibility criteria and were included. Dentin characteristics and mechanical properties alter with age, and spatially within a tooth to depend on tubule orientation. Age-related mineral deposition within tubules, and collagen maturation in intertubular dentin compound the spatial effects on mechanical properties. Mechanical properties depend on collagen fiber orientation and apatite alignment relative to dentin tubules, characteristic differences in peritubular and intertubular dentin, and relative dentin tubule distribution within a tooth. The bulk of the research focussed on age-related apatite effects, although many reported limited understanding of changes to collagen, particularly in intertubular dentin. CONCLUSION: Investigations into the effect of age, depth, site and location on dentin collagen are warranted to minimize tooth loss in older populations by providing targeted adhesive, restorative or preventative interventions.


Asunto(s)
Dentina , Diente , Adulto , Humanos , Anciano , Colágeno , Apatitas , Minerales
12.
Int J Mol Sci ; 23(17)2022 Aug 26.
Artículo en Inglés | MEDLINE | ID: covidwho-2023746

RESUMEN

Although interstitial lung disease (ILD) is a life-threatening pathological condition that causes respiratory failure, the efficiency of current therapies is limited. This study aimed to investigate the effects of human MIKO-1 (hMIKO-1), a hybrid protein that suppresses the abnormal activation of macrophages, on murine macrophage function and its therapeutic effect in a mouse model of bleomycin-induced ILD (BLM-ILD). To this end, the phenotype of thioglycolate-induced murine peritoneal macrophages co-cultured with hMIKO-1 was examined. The mice were assigned to normal, BLM-alone, or BLM + hMIKO-1 groups, and hMIKO-1 (0.1 mg/mouse) was administered intraperitoneally from day 0 to 14. The mice were sacrificed on day 28, and their lungs were evaluated by histological examination, collagen content, and gene expression levels. hMIKO-1 suppressed the polarization of murine macrophages to M2 predominance in vitro. The fibrosis score of lung pathology and lung collagen content of the BLM + hMIKO-1 group were significantly lower than those in the BLM-alone group. The expression levels of TNF-α, IL-6, IL-1ß, F4/80, and TIMP-1 in the lungs of the BLM + hMIKO-1 group were significantly lower than those in the BLM-alone group. These findings indicate that hMIKO-1 reduces lung fibrosis and may be a future therapeutic candidate for ILD treatment.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Fibrosis Pulmonar , Animales , Bleomicina/toxicidad , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Pulmón/patología , Enfermedades Pulmonares Intersticiales/inducido químicamente , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo
13.
Peptides ; 157: 170848, 2022 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1967000

RESUMEN

Angiotensin (Ang) II, the main active member of the renin angiotensin system (RAS), is essential for the maintenance of cardiovascular homeostasis. However, hyperactivation of the RAS causes fibrotic diseases. Ang II has pro-inflammatory actions, and moreover activates interstitial fibroblasts and/or dysregulates extracellular matrix degradation. The discovery of new RAS pathways has revealed the complexity of this system. Among the RAS peptides, alamandine (ALA, Ala1 Ang 1-7) has been identified in humans, rats, and mice, with protective actions in different pathological conditions. ALA has similar effects to its well-known congener, Ang-(1-7), as a vasodilator, anti-inflammatory, and antifibrotic. Its protective role against cardiovascular diseases is well-reviewed in the literature. However, the protective actions of ALA in fibrotic conditions have been little explored. Therefore, in this article, we review the ability of ALA to modulate the inflammatory process and collagen deposition, to serve as an antioxidant, and to mediate protection against functional disorders. In this scenario, we also explore ALA as a promising therapy for pulmonary fibrosis after COVID-19 infection.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Peptidil-Dipeptidasa A , Angiotensina II/metabolismo , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antioxidantes/farmacología , Colágeno/metabolismo , Fibrosis , Humanos , Ratones , Oligopéptidos , Peptidil-Dipeptidasa A/metabolismo , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina , Vasodilatadores/farmacología
14.
J Thromb Thrombolysis ; 54(2): 211-216, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-1959071

RESUMEN

The COVID-19 pandemic is often accompanied by severe respiratory illness and thrombotic complications. Von Willebrand Factor (VWF) levels are highly elevated in this condition. However, limited data are available on the qualitative activity of VWF in COVID-19. We measured plasma VWF levels quantitatively (VWF antigen) and qualitatively (ristocetin-induced platelet agglutination, glycoprotein IbM (GPIbM) binding, and collagen binding). Consistent with prior reports, VWF antigen levels were significantly elevated in hospitalized patients with or without COVID-19. The GPIbM and collagen binding activity-to-antigen ratios were significantly reduced, consistent with qualitative changes in VWF in COVID-19. Of note, critically ill hospitalized patients without COVID-19 had similar reductions in VWF activity-to-antigen ratios as patients with COVID-19. Our data suggest that qualitative changes in VWF in COVID-19 may not be specific to COVID-19. Future studies are warranted to determine the mechanisms responsible for qualitative changes in VWF in COVID-19 and other critical illnesses.• VWF levels were increased in COVID-19 compared to healthy controls.• VWF activity-to-antigen ratios were decreased in COVID-19 compared to healthy controls.• There were no differences in VWF activity-to-antigen ratios between hospitalized patients with or without COVID-19.• These findings are consistent with qualitative changes in VWF in systemic inflammation which are not specific to COVID-19.• Future studies are needed to define possible roles of changes in conformation or multimer length in the qualitative changes in VWF in systemic inflammation.


Asunto(s)
COVID-19 , Enfermedades de von Willebrand , Colágeno , Humanos , Inflamación , Pandemias , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Factor de von Willebrand/metabolismo
15.
Genes (Basel) ; 13(5)2022 04 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1875530

RESUMEN

We investigated four cats with similar clinical skin-related signs strongly suggestive of Ehlers-Danlos syndrome (EDS). Cases no. 1 and 4 were unrelated and the remaining two cases, no. 2 and 3, were reportedly siblings. Histopathological changes were characterized by severely altered dermal collagen fibers. Transmission electron microscopy in one case demonstrated abnormalities in the collagen fibril organization and structure. The genomes of the two unrelated affected cats and one of the affected siblings were sequenced and individually compared to 54 feline control genomes. We searched for private protein changing variants in known human EDS candidate genes and identified three independent heterozygous COL5A1 variants. COL5A1 is a well-characterized candidate gene for classical EDS. It encodes the proα1 chain of type V collagen, which is needed for correct collagen fibril formation and the integrity of the skin. The identified variants in COL5A1 are c.112_118+15del or r.spl?, c.3514A>T or p.(Lys1172*), and c.3066del or p.(Gly1023Valfs*50) for cases no. 1, 2&3, and 4, respectively. They presumably all lead to nonsense-mediated mRNA decay, which results in haploinsufficiency of COL5A1 and causes the alterations of the connective tissue. The whole genome sequencing approach used in this study enables a refinement of the diagnosis for the affected cats as classical EDS. It further illustrates the potential of such experiments as a precision medicine approach in animals with inherited diseases.


Asunto(s)
Síndrome de Ehlers-Danlos , Animales , Gatos/genética , Colágeno/genética , Colágeno Tipo V/genética , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/veterinaria , Exones
16.
Clin Transl Med ; 12(5): e831, 2022 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1858584

RESUMEN

Tissue damage caused by an infection oran autoimmune disease triggers degradation of collagen in the extracellular matrix (ECM), which further enhances inflammation. Therefore, improving ECM in aninflamed tissue can be exploited as a potential therapeutic target. A recentstudy emphasised an innovative approach against COVID-19 using polymerised type I collagen (PTIC) that improves disease severity through a hitherto unknownmechanism. In this paper, we provide an overview of potential mechanism thatmay explain the anti-inflammatory effect of collagen peptides. In addition,the paper includes a brief summary of possible side effect of collagendeposition in inflammatory diseases. Altogether, current knowledge suggeststhat collagen may potentially reduce the residual risk in inflammatorydiseases; however, the detailed mechanism remains elusive.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Colágeno/metabolismo , Colágeno/farmacología , Colágeno Tipo I/metabolismo , Colágeno Tipo I/farmacología , Matriz Extracelular/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo
17.
Cells ; 11(5)2022 03 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1742340

RESUMEN

Mast cells are widely distributed in various parts of the human body and play a vital role in the progression of many diseases. Recently, the close relationship between mast cells and acupoints was elucidated, and the role of mast cells in acupuncture analgesia has attracted the attention of researchers worldwide. Using mast cells, acupuncture analgesia and acupoint as key words to search CNKI, PubMed, Web of Science and other databases, combining the representative articles in these databases with the published research papers of our group, we summarized: The enrichment of mast cells and the dense arrangement of collagen fibers, microvessels, and nerves form the basis for acupoints as the reaction sites of acupuncture; acupuncture can cause the deformation of collagen fibers and activate TRPV channels on mast cells membrane, so as to stimulate mast cells to release bioactive substances and activate nerve receptors to generate analgesic effect; system biology models are set up to explain the quantitative process of information initiation and transmission at acupuncture points, and indicate that the acupuncture effect depends on the local mast cells density. In a conclusion, this review will give a scientific explanation of acupuncture analgesia from the material basis of acupoints, the local initiation, and afferent biological mechanism.


Asunto(s)
Analgesia por Acupuntura , Terapia por Acupuntura , Puntos de Acupuntura , Colágeno , Humanos , Mastocitos/fisiología
18.
J Cosmet Dermatol ; 21(5): 1816-1821, 2022 May.
Artículo en Inglés | MEDLINE | ID: covidwho-1714236

RESUMEN

BACKGROUND: Acute inflammatory reactions (AIRs) are a rare complication following esthetic treatment with hyaluronic acid (HA) and/or human collagen fillers. However, a substantial increase in the frequency of AIRs was observed in the first author's clinic since May 2020. AIMS: To report AIR cases, we experienced and discuss potential underlying mechanisms. METHODS: This was a retrospective review of patients representing AIR symptoms following filler injection with HA or human collagen in our clinic. RESULTS: Although only one case of an AIR with an incidence rate of 0.01% was recorded following filler treatment between September 2008 and April 2020 in our clinic, we observed 14 AIR cases without anaphylaxis, with an incidence rate of 1.18% between May 2020 and June 2021, in line with the spreading of the new coronavirus pandemic. All cases were females aged 40-57 years, and the time of onset was within hours after filler injection. Three patients had been treated with HA fillers only, 2 with HA plus human collagen, and 9 with human collagen only. Most patients had been treated with these products in the past. Nine patients were treated with oral prednisolone. In all cases, symptoms resolved entirely within a week without sequelae. CONCLUSIONS: The marked increase in AIRs coincided with the COVID-19 pandemic. Possible explanations include immune system alterations caused by extensive changes in domestic and personal hygiene, prolonged and elevated stress levels, and subclinical COVID-19 infection. Further studies may be warranted.


Asunto(s)
COVID-19 , Técnicas Cosméticas , Rellenos Dérmicos , COVID-19/epidemiología , Colágeno/efectos adversos , Técnicas Cosméticas/efectos adversos , Rellenos Dérmicos/efectos adversos , Femenino , Humanos , Ácido Hialurónico/efectos adversos , Incidencia , Masculino , Pandemias
19.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1631216

RESUMEN

Angiotensin II receptor type 1 (AT1R) and endothelin-1 receptor type A (ETAR) are G-protein-coupled receptors (GPCRs) expressed on the surface of a great variety of cells: immune cells, vascular smooth cells, endothelial cells, and fibroblasts express ETAR and AT1R, which are activated by endothelin 1 (ET1) and angiotensin II (AngII), respectively. Certain autoantibodies are specific for these receptors and can regulate their function, thus being known as functional autoantibodies. The function of these antibodies is similar to that of natural ligands, and it involves not only vasoconstriction, but also the secretion of proinflammatory cytokines (such as interleukin-6 (IL6), IL8 and TNF-α), collagen production by fibroblasts, and reactive oxygen species (ROS) release by fibroblasts and neutrophils. The role of autoantibodies against AT1R and ETAR (AT1R-AAs and ETAR-AAs, respectively) is well described in the pathogenesis of many medical conditions (e.g., systemic sclerosis (SSc) and SSc-associated pulmonary hypertension, cystic fibrosis, and allograft dysfunction), but their implications in cardiovascular diseases are still unclear. This review summarizes the current evidence regarding the effects of AT1R-AAs and ETAR-AAs in cardiovascular pathologies, highlighting their roles in heart transplantation and mechanical circulatory support, preeclampsia, and acute coronary syndromes.


Asunto(s)
Autoanticuerpos/metabolismo , Enfermedades Cardiovasculares/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Receptor de Endotelina A/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Colágeno/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Horm Mol Biol Clin Investig ; 43(3): 353-355, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1573640

RESUMEN

OBJECTIVES: To evaluate the potential relationship between COVID-19 pandemic and mucormycosis outbreak. METHODS: PubMed, Embase, Cochrane Library and Google Scholar were searched for the term "COVID-19 and mucormycosis" up to May 31, 2021. RESULTS: After the second wave of COVID-19, the mucormycosis outbreak complicates the natural course of COVID-19. COVID-19 patients with uncontrolled diabetes mellitus with diabetic ketoacidosis, excessive glucocorticoid use, prolonged neutropenia, malnutrition and any underlying immunocompromised conditions are at risk of developing mucormycosis. CONCLUSIONS: Hyperglycaemia impairs the motility of phagocytes and also decreases the oxidative and non-oxidative mechanism of killing the causative pathogen. Chronic hyperglycemia also leads to the formation of advanced glycation end-products (AGE), which leads to cross-linking between key proteins of inflammation and connective tissue such as collagen which makes tissue susceptible to immunological dysregulation. The receptor for AGE (RAGE) is expressed on various inflammatory cells including neutrophils and its activation by AGEs leads to activation of many down signaling pathways which ultimately leads to impairment of the inflammatory response. Hyperglycemia also increases serum Nitric Oxide (NO), which decreases neutrophil motility and reduces the synthesis and release of various inflammatory mediators such as TNF-α and IL-1ß, IL-6. It also decreases the expression of adhesion molecules such as LFA-1 and ICAM-2, on neutrophils. Steroids cause immunosuppression majorly by inhibiting the NF-κB pathway which is a transcription factor involved in the synthesis of many immunological mediators such as Interleukins, cytokines, chemokines, etc., and various adhesion molecules.


Asunto(s)
COVID-19 , Diabetes Mellitus , Hiperglucemia , Mucormicosis , COVID-19/complicaciones , Colágeno , Citocinas/metabolismo , Diabetes Mellitus/epidemiología , Glucocorticoides , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Hiperglucemia/epidemiología , Mediadores de Inflamación , Interleucina-6 , Antígeno-1 Asociado a Función de Linfocito , Mucormicosis/epidemiología , FN-kappa B/metabolismo , Óxido Nítrico , Pandemias , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Factor de Necrosis Tumoral alfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA