Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Braz. J. Pharm. Sci. (Online) ; 59: e20200, 2023. graf
Artículo en Inglés | LILACS | ID: biblio-1505842

RESUMEN

Abstract The renin-angiotensin-aldosterone system (RAAS) plays a key role in diabetic nephropathy (DN). Angiotensin-II secreted during the RAAS pathway increases nephropathy. It stimulates oxidative stress which can quench nitric oxide. Reduced nitric oxide level aggravates Ang-II-induced vasoconstriction. Ang-II has also emerged as a central mediator of the glomerular hemodynamic changes that are associated with renal injury. Deletion of ACE2 is also noted due to increased Ang-II level which leads to the development of DN. We hypothesize that nephropathy caused by Ang-II in the periphery may be controlled by brain RAAS. ACE inhibitors and ARBs may show the renoprotective effect when administered through ICV without crossing the blood-brain barrier. DN was observed after 8 weeks of diabetes induction through alloxan. Administration of captopril and valsartan once and in combined therapy for 2 weeks, significantly reduced urine output, blood urea nitrogen, total protein in the urine, serum cholesterol, serum creatinine, serum triglycerides, and kidney/body weight ratio as compared to diabetic control rats. Further, combination therapy significantly increased the body weight and serum nitrate level as compared to diabetic control animals. However, increased ACE2 levels in the brain may reduce the sympathetic outflow and might have decreased the peripheral activity of Ang-II which shows beneficial effects in DN.


Asunto(s)
Animales , Masculino , Femenino , Ratas , Sistema Renina-Angiotensina/inmunología , Angiotensina II/análisis , Nefropatías Diabéticas/patología , Heridas y Lesiones/clasificación , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Peptidil-Dipeptidasa A/administración & dosificación
2.
Nat Commun ; 13(1): 1220, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35264564

RESUMEN

COVID-19 shares the feature of autoantibody production with systemic autoimmune diseases. In order to understand the role of these immune globulins in the pathogenesis of the disease, it is important to explore the autoantibody spectra. Here we show, by a cross-sectional study of 246 individuals, that autoantibodies targeting G protein-coupled receptors (GPCR) and RAS-related molecules associate with the clinical severity of COVID-19. Patients with moderate and severe disease are characterized by higher autoantibody levels than healthy controls and those with mild COVID-19 disease. Among the anti-GPCR autoantibodies, machine learning classification identifies the chemokine receptor CXCR3 and the RAS-related molecule AGTR1 as targets for antibodies with the strongest association to disease severity. Besides antibody levels, autoantibody network signatures are also changing in patients with intermediate or high disease severity. Although our current and previous studies identify anti-GPCR antibodies as natural components of human biology, their production is deregulated in COVID-19 and their level and pattern alterations might predict COVID-19 disease severity.


Asunto(s)
Autoanticuerpos/inmunología , COVID-19/inmunología , Receptores Acoplados a Proteínas G/inmunología , Sistema Renina-Angiotensina/inmunología , Autoanticuerpos/sangre , Autoinmunidad , Biomarcadores/sangre , COVID-19/sangre , COVID-19/clasificación , Estudios Transversales , Femenino , Humanos , Aprendizaje Automático , Masculino , Análisis Multivariante , Receptor de Angiotensina Tipo 1/inmunología , Receptores CXCR3/inmunología , SARS-CoV-2 , Índice de Severidad de la Enfermedad
3.
Signal Transduct Target Ther ; 7(1): 57, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197452

RESUMEN

The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.


Asunto(s)
Lesión Renal Aguda/complicaciones , COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/complicaciones , Coagulación Intravascular Diseminada/complicaciones , Linfopenia/complicaciones , Miocarditis/complicaciones , Embolia Pulmonar/complicaciones , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/virología , Anticoagulantes/uso terapéutico , Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/virología , Ensayos Clínicos como Asunto , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Coagulación Intravascular Diseminada/tratamiento farmacológico , Coagulación Intravascular Diseminada/inmunología , Coagulación Intravascular Diseminada/virología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/virología , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Linfopenia/tratamiento farmacológico , Linfopenia/inmunología , Linfopenia/virología , Miocarditis/tratamiento farmacológico , Miocarditis/inmunología , Miocarditis/virología , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/inmunología , Embolia Pulmonar/virología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Tratamiento Farmacológico de COVID-19
4.
Brain Res ; 1772: 147667, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34587500

RESUMEN

INTRODUCTION: As several clinical trials have revealed that angiotensin-converting enzyme inhibitors and angiotensin II (Ang II) receptor blockers may be efficient in treating vascular dementia (VaD), the long-acting blockade of the renin-angiotensin system (RAS) would be useful considering the poor adherence of antihypertensive drugs. Accordingly, we continuously blocked RAS via vaccination and examined the effectiveness of the VaD model in rats. METHODS: Male Wistar rats were exposed to two-vessel occlusions (2VO) after three injections of Ang II peptide vaccine. The effects of the vaccine were evaluated in the novel object recognition test, brain RAS components, and markers for oligodendrocytes. RESULTS: In the vaccinated rats, anti-Ang II antibody titer level was increased in serum until Day 168, but not in cerebral parenchyma. Vaccinated rats showed better object recognition memory with inhibited demyelination in the corpus callosum and activation of astrocytes and microglia. Also, levels of BrdU/GSTπ-positive cells and the phosphorylation of cAMP response element binding protein was increased in vaccinated rats, indicating that the differentiation of oligodendrocyte progenitor cells to mature oligodendrocytes was accelerated. Vaccinated rats showed increased expression of fibroblast growth factor-2 (FGF2), which was observed in endothelial cells. Angiotensinogen mRNA was decreased at 7 days after 2VO but increased at 14 and 28 days. CONCLUSION: Ang II vaccine might have promoted oligodendrocyte differentiation and inhibited astrocytic and microglial activation by stimulating FGF2 signaling in the endothelial cells-oligodendrocyte/astrocyte/microglia coupling. These data indicate the feasibility of Ang II vaccine for preventing progression of vascular dementia.


Asunto(s)
Demencia Vascular/prevención & control , Inmunoterapia/métodos , Sistema Renina-Angiotensina/inmunología , Angiotensina II/inmunología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Anticuerpos/análisis , Encéfalo/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Enfermedades Desmielinizantes/prevención & control , Masculino , Memoria/fisiología , Fosforilación , Ratas , Ratas Wistar , Reconocimiento en Psicología , Vacunación , Vacunas de Subunidad/uso terapéutico
5.
PLoS One ; 16(9): e0257016, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34478478

RESUMEN

BACKGROUND: Activation of the immune system is implicated in the Post-Acute Sequelae after SARS-CoV-2 infection (PASC) but the mechanisms remain unknown. Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II (Ang II) resulting in decreased activation of the AT1 receptor and decreased immune system activation. We hypothesized that autoantibodies against ACE2 may develop after SARS-CoV-2 infection, as anti-idiotypic antibodies to anti-spike protein antibodies. METHODS AND FINDINGS: We tested plasma or serum for ACE2 antibodies in 67 patients with known SARS-CoV-2 infection and 13 with no history of infection. None of the 13 patients without history of SARS-CoV-2 infection and 1 of the 20 outpatients that had a positive PCR test for SARS-CoV-2 had levels of ACE2 antibodies above the cutoff threshold. In contrast, 26/32 (81%) in the convalescent group and 14/15 (93%) of patients acutely hospitalized had detectable ACE2 antibodies. Plasma from patients with antibodies against ACE2 had less soluble ACE2 activity in plasma but similar amounts of ACE2 protein compared to patients without ACE2 antibodies. We measured the capacity of the samples to inhibit ACE2 enzyme activity. Addition of plasma from patients with ACE2 antibodies led to decreased activity of an exogenous preparation of ACE2 compared to patients that did not have antibodies. CONCLUSIONS: Many patients with a history of SARS-CoV-2 infection have antibodies specific for ACE2. Patients with ACE2 antibodies have lower activity of soluble ACE2 in plasma. Plasma from these patients also inhibits exogenous ACE2 activity. These findings are consistent with the hypothesis that ACE2 antibodies develop after SARS-CoV-2 infection and decrease ACE2 activity. This could lead to an increase in the abundance of Ang II, which causes a proinflammatory state that triggers symptoms of PASC.


Asunto(s)
Autoanticuerpos/sangre , COVID-19/inmunología , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/sangre , Angiotensina II/sangre , Angiotensina II/inmunología , Enzima Convertidora de Angiotensina 2/genética , Autoanticuerpos/inmunología , Autoanticuerpos/aislamiento & purificación , COVID-19/sangre , COVID-19/virología , Femenino , Humanos , Masculino , Peptidil-Dipeptidasa A/sangre , Receptor de Angiotensina Tipo 1/sangre , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/inmunología , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/aislamiento & purificación
6.
Cells ; 10(7)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34359936

RESUMEN

The renin-angiotensin system (RAS) has long been described in the field of cardiovascular physiology as the main player in blood pressure homeostasis. However, other effects have since been described, and include proliferation, fibrosis, and inflammation. To illustrate the immunomodulatory properties of the RAS, we chose three distinct fields in which RAS may play a critical role and be the subject of specific treatments. In oncology, RAS hyperactivation has been associated with tumor migration, survival, cell proliferation, and angiogenesis; preliminary data showed promise of the benefit of RAS blockers in patients treated for certain types of cancer. In intensive care medicine, vasoplegic shock has been associated with severe macro- and microcirculatory imbalance. A relative insufficiency in angiotensin II (AngII) was associated to lethal outcomes and synthetic AngII has been suggested as a specific treatment in these cases. Finally, in solid organ transplantation, both AngI and AngII have been associated with increased rejection events, with a regional specificity in the RAS activity. These elements emphasize the complexity of the direct and indirect interactions of RAS with immunomodulatory pathways and warrant further research in the field.


Asunto(s)
Inmunomodulación , Sistema Renina-Angiotensina/inmunología , Animales , Humanos , Unidades de Cuidados Intensivos , Neoplasias/metabolismo , Transducción de Señal , Trasplante
7.
Int Immunopharmacol ; 98: 107913, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34218218

RESUMEN

The objective of this work was to study the effects and mechanisms of S-allylmercapto-N-acetylcysteine (ASSNAC) in the treatment of pulmonary emphysema based on network pharmacology analysis and other techniques. Firstly, the potential targets associated with ASSNAC and COPD were integrated using public databases. Then, a protein-protein interaction network was constructed using String database and Cytoscape software. The Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed on DAVID platform. The molecular docking of ASSNAC with some key disease targets was implemented on the SwissDock platform. To verify the results of the network pharmacology, a pulmonary emphysema mice model was established and treated with ASSNAC. Besides, the expressions of the predicted targets were detected by immunohistochemistry, Western blot analysis or enzyme-linked immunosorbent assay. Results showed that 33 overlapping targets are achieved, including CXCL8, ICAM1, MAP2K1, PTGS2, ACE and so on. The critical pathways of ASSNAC against COPD involved arachidonic acid metabolism, chemokine pathway, MAPK pathway, renin-angiotensin system, and others. Pharmacodynamic experiments demonstrated that ASSNAC decreased the pulmonary emphysema and inflammation in the pulmonary emphysema mice. Therefore, these results confirm the perspective of network pharmacology in the target verification, and indicate the treatment potential of ASSNAC against COPD.


Asunto(s)
Acetilcisteína/análogos & derivados , Compuestos Alílicos/farmacología , Antiinflamatorios/farmacología , Enfisema Pulmonar/tratamiento farmacológico , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Compuestos Alílicos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Farmacología en Red , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/inmunología , Enfisema Pulmonar/diagnóstico , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
8.
Front Immunol ; 12: 686029, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276671

RESUMEN

More than one year since its emergence, corona virus disease 2019 (COVID-19) is still looming large with a paucity of treatment options. To add to this burden, a sizeable subset of patients who have recovered from acute COVID-19 infection have reported lingering symptoms, leading to significant disability and impairment of their daily life activities. These patients are considered to suffer from what has been termed as "chronic" or "long" COVID-19 or a form of post-acute sequelae of COVID-19, and patients experiencing this syndrome have been termed COVID-19 long-haulers. Despite recovery from infection, the persistence of atypical chronic symptoms, including extreme fatigue, shortness of breath, joint pains, brain fogs, anxiety and depression, that could last for months implies an underlying disease pathology that persist beyond the acute presentation of the disease. As opposed to the direct effects of the virus itself, the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is believed to be largely responsible for the appearance of these lasting symptoms, possibly through facilitating an ongoing inflammatory process. In this review, we hypothesize potential immunological mechanisms underlying these persistent and prolonged effects, and describe the multi-organ long-term manifestations of COVID-19.


Asunto(s)
COVID-19/complicaciones , SARS-CoV-2/inmunología , Ansiedad/etiología , Artralgia/etiología , Autoinmunidad , COVID-19/epidemiología , COVID-19/inmunología , COVID-19/virología , Depresión/etiología , Disnea/etiología , Fatiga/etiología , Microbioma Gastrointestinal/inmunología , Humanos , Huésped Inmunocomprometido , Incidencia , Prevalencia , Sistema Renina-Angiotensina/inmunología , Síndrome Post Agudo de COVID-19
9.
J Histochem Cytochem ; 69(12): 835-847, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34165363

RESUMEN

Cancer stem cells (CSCs) drive metastasis, treatment resistance, and tumor recurrence. CSCs reside within a niche, an anatomically distinct site within the tumor microenvironment (TME) that consists of malignant and non-malignant cells, including immune cells. The renin-angiotensin system (RAS), a critical regulator of stem cells and key developmental processes, plays a vital role in the TME. Non-malignant cells within the CSC niche and stem cell signaling pathways such as the Wnt, Hedgehog, and Notch pathways influence CSCs. Components of the RAS and cathepsins B and D that constitute bypass loops of the RAS are expressed on CSCs in many cancer types. There is extensive in vitro and in vivo evidence showing that RAS inhibition reduces tumor growth, cell proliferation, invasion, and metastasis. However, there is inconsistent epidemiological data on the effect of RAS inhibitors on cancer incidence and survival outcomes, attributed to different patient characteristics and methodologies used between studies. Further mechanistic studies are warranted to investigate the precise effects of the RAS on CSCs directly and/or the CSC niche. Targeting the RAS, its bypass loops, and convergent signaling pathways participating in the TME and other key stem cell pathways that regulate CSCs may be a novel approach to cancer treatment.


Asunto(s)
Neoplasias/terapia , Células Madre Neoplásicas/metabolismo , Sistema Renina-Angiotensina/inmunología , Nicho de Células Madre/inmunología , Animales , Catepsinas/inmunología , Proliferación Celular , Reposicionamiento de Medicamentos , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Humanos , Transducción de Señal , Microambiente Tumoral
10.
J Autoimmun ; 122: 102683, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34144328

RESUMEN

The renin-angiotensin system (RAS) plays a major role in COVID-19. Severity of several inflammation-related diseases has been associated with autoantibodies against RAS, particularly agonistic autoantibodies for angiotensin type-1 receptors (AA-AT1) and autoantibodies against ACE2 (AA-ACE2). Disease severity of COVID-19 patients was defined as mild, moderate or severe following the WHO Clinical Progression Scale and determined at medical discharge. Serum AA-AT1 and AA-ACE2 were measured in COVID-19 patients (n = 119) and non-infected controls (n = 23) using specific solid-phase, sandwich enzyme-linked immunosorbent assays. Serum LIGHT (TNFSF14; tumor necrosis factor ligand superfamily member 14) levels were measured with the corresponding assay kit. At diagnosis, AA-AT1 and AA-ACE2 levels were significantly higher in the COVID-19 group relative to controls, and we observed significant association between disease outcome and serum AA-AT1 and AA-ACE2 levels. Mild disease patients had significantly lower levels of AA-AT1 (p < 0.01) and AA-ACE2 (p < 0.001) than moderate and severe patients. No significant differences were detected between males and females. The increase in autoantibodies was not related to comorbidities potentially affecting COVID-19 severity. There was significant positive correlation between serum levels of AA-AT1 and LIGHT (TNFSF14; rPearson = 0.70, p < 0.001). Both AA-AT1 (by agonistic stimulation of AT1 receptors) and AA-ACE2 (by reducing conversion of Angiotensin II into Angiotensin 1-7) may lead to increase in AT1 receptor activity, enhance proinflammatory responses and severity of COVID-19 outcome. Patients with high levels of autoantibodies require more cautious control after diagnosis. Additionally, the results encourage further studies on the possible protective treatment with AT1 receptor blockers in COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/inmunología , Autoanticuerpos/sangre , Autoantígenos/inmunología , COVID-19/inmunología , Receptor de Angiotensina Tipo 1/inmunología , Anciano , Autoanticuerpos/inmunología , COVID-19/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2
11.
Mol Cell Endocrinol ; 529: 111254, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33798633

RESUMEN

The most classical view of the renin-angiotensin system (RAS) emphasizes its role as an endocrine regulator of sodium balance and blood pressure. However, it has long become clear that the RAS has pleiotropic actions that contribute to organ damage, including modulation of inflammation. Angiotensin II (Ang II) activates angiotensin type 1 receptors (AT1R) to promote an inflammatory response and organ damage. This represents the pathophysiological basis for the successful use of RAS blockers to prevent and treat kidney and heart disease. However, other RAS components could have a built-in capacity to brake proinflammatory responses. Angiotensin type 2 receptor (AT2R) activation can oppose AT1R actions, such as vasodilatation, but its involvement in modulation of inflammation has not been conclusively proven. Angiotensin-converting enzyme 2 (ACE2) can process Ang II to generate angiotensin-(1-7) (Ang-(1-7)), that activates the Mas receptor to exert predominantly anti-inflammatory responses depending on the context. We now review recent advances in the understanding of the interaction of the RAS with inflammation. Specific topics in which novel information became available recently include intracellular angiotensin receptors; AT1R posttranslational modifications by tissue transglutaminase (TG2) and anti-AT1R autoimmunity; RAS modulation of lymphoid vessels and T lymphocyte responses, especially of Th17 and Treg responses; interactions with toll-like receptors (TLRs), programmed necrosis, and regulation of epigenetic modulators (e.g. microRNAs and bromodomain and extraterminal domain (BET) proteins). We additionally discuss an often overlooked effect of the RAS on inflammation which is the downregulation of anti-inflammatory factors such as klotho, peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), transient receptor potential ankyrin 1 (TRPA1), SNF-related serine/threonine-protein kinase (SNRK), serine/threonine-protein phosphatase 6 catalytic subunit (Ppp6C) and n-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). Both transcription factors, such as nuclear factor κB (NF-κB), and epigenetic regulators, such as miRNAs are involved in downmodulation of anti-inflammatory responses. A detailed analysis of pathways and targets for downmodulation of anti-inflammatory responses constitutes a novel frontier in RAS research.


Asunto(s)
Angiotensina II/inmunología , Angiotensina I/inmunología , Inflamación/inmunología , Fragmentos de Péptidos/inmunología , Sistema Renina-Angiotensina/inmunología , Equilibrio Hidroelectrolítico/inmunología , Angiotensina I/genética , Angiotensina II/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Animales , Autoinmunidad , Presión Sanguínea/genética , Presión Sanguínea/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Riñón/citología , Riñón/inmunología , Proteínas Klotho/genética , Proteínas Klotho/inmunología , Fragmentos de Péptidos/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/inmunología , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/inmunología , Receptor de Angiotensina Tipo 2/genética , Receptor de Angiotensina Tipo 2/inmunología , Sistema Renina-Angiotensina/genética , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología , Equilibrio Hidroelectrolítico/genética
13.
Mol Biol Rep ; 48(3): 2917-2928, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33837899

RESUMEN

The renin-angiotensin-aldosterone system and its metabolites play an important role in homeostasis of body, especially the cardiovascular system. In this study, we discuss the imbalance of multiple systems during the infection and the importance of therapeutic choice, dosing, and laboratory monitoring of cardiac and anti-coagulant therapies in COVID-19 patients. The crosstalk between angiotensin, kinin-kallikrein system, as well as inflammatory and coagulation systems plays an essential role in COVID-19. Cardiac complications and coagulopathies imply the crosstalks between the mentioned systems. We believe that the blockage of bradykinin can be a good option in the management of COVID-19 and CVD in patients and that supportive treatment of respiratory and cardiologic complications is needed in COVID-19 patients. Ninety-one percent of COVID-19 patients who were admitted to hospital with a prolonged aPTT were positive for lupus anticoagulant, which increases the risk of thrombosis and prolonged aPTT. Therefore, the question that is posed at this juncture is whether it is safe to use the prophylactic dose of heparin particularly in those with elevated D-dimer levels. It should be noted that timing is of high importance in anti-coagulant therapy; therefore, we should consider the level of D-dimer, fibrinogen, drug-drug interactions, and risk factors during thromboprophylaxis administration. Fibrinogen is an independent predictor of resistance to heparin and should be considered before thromboprophylaxis. Alteplase and Futhan might be a good choice to assess the condition of heparin resistance. Finally, the treatment option, dosing, and laboratory monitoring of anticoagulant therapy are critical decisions in COVID-19 patients.


Asunto(s)
COVID-19 , SARS-CoV-2 , Trombosis , Anticoagulantes/administración & dosificación , Anticoagulantes/uso terapéutico , Bradiquinina/sangre , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/fisiopatología , COVID-19/terapia , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Humanos , Inflamación/inmunología , Inflamación/virología , Calicreínas/sangre , Sistema Renina-Angiotensina/inmunología , Sistema Renina-Angiotensina/fisiología , Trombosis/tratamiento farmacológico , Trombosis/prevención & control , Trombosis/virología
14.
J Clin Pharmacol ; 61(8): 987-1000, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33635546

RESUMEN

Since the discovery of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), numerous research has been undertaken to delineate the various effects of the virus which manifests in many ways all over the body. The association between the SARS-CoV-2 invasion mechanism and the renin-angiotensin-aldosterone system (RAAS) receptors, created many debates about the possible consequences of using RAAS-modulating drugs including angiotensin-converting enzyme inhibitors (ACEi) and angiotensin II receptor blockers (ARBs) during the pandemic. Many clinical studies were conducted to assess the outcomes of coronavirus disease 2019 (COVID-19) in patients who use ACEi/ARBs following the arguments claiming to discontinue these drugs as a precautionary measure. Although several studies mainly analyzed the outcomes of the disease, this review aimed to compare specific blood markers in both groups of COVID-19 patients to gain better insight into the interaction of ACEi/ARBs with different body functions during the infection. Several databases were searched using a combination of keywords followed by screening and data extraction. Only 28 studies met our inclusion criteria, the majority of which showed no significant difference between the inflammation markers of COVID-19 patients who used or did not use ACEi/ARBs. Interestingly, 6 studies reported lower inflammatory markers in COVID-19 patients who used ACEi/ARBs, and 6 studies reported better outcomes among the same group. We therefore concluded that the use of ACEi/ARBs may not lead to worse prognosis of COVID-19 and may even play a protective role against the hyperinflammatory response associated with COVID-19.


Asunto(s)
Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , COVID-19 , Inmunidad , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2/fisiología , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/metabolismo , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/fisiología , Pronóstico , Factores Protectores
15.
J Neurovirol ; 27(1): 35-51, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33534131

RESUMEN

Since the outbreak of coronavirus disease 2019 (COVID-19) in 2019, it is gaining worldwide attention at the moment. Apart from respiratory manifestations, neurological dysfunction in COVID-19 patients, especially the occurrence of cerebrovascular diseases (CVD), has been intensively investigated. In this review, the effects of COVID-19 infection on CVD were summarized as follows: (I) angiotensin-converting enzyme 2 (ACE2) may be involved in the attack on vascular endothelial cells by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), leading to endothelial damage and increased subintimal inflammation, which are followed by hemorrhage or thrombosis; (II) SARS-CoV-2 could alter the expression/activity of ACE2, consequently resulting in the disruption of renin-angiotensin system which is associated with the occurrence and progression of atherosclerosis; (III) upregulation of neutrophil extracellular traps has been detected in COVID-19 patients, which is closely associated with immunothrombosis; (IV) the inflammatory cascade induced by SARS-CoV-2 often leads to hypercoagulability and promotes the formation and progress of atherosclerosis; (V) antiphospholipid antibodies are also detected in plasma of some severe cases, which aggravate the thrombosis through the formation of immune complexes; (VI) hyperglycemia in COVID-19 patients may trigger CVD by increasing oxidative stress and blood viscosity; (VII) the COVID-19 outbreak is a global emergency and causes psychological stress, which could be a potential risk factor of CVD as coagulation, and fibrinolysis may be affected. In this review, we aimed to further our understanding of CVD-associated COVID-19 infection, which could improve the therapeutic outcomes of patients. Personalized treatments should be offered to COVID-19 patients at greater risk for stroke in future clinical practice.


Asunto(s)
Aterosclerosis/complicaciones , COVID-19/complicaciones , Coagulación Intravascular Diseminada/complicaciones , Hemorragia/complicaciones , Hiperglucemia/complicaciones , Accidente Cerebrovascular/complicaciones , Trombosis/complicaciones , Anticoagulantes/uso terapéutico , Antivirales/uso terapéutico , Aterosclerosis/diagnóstico , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/virología , COVID-19/diagnóstico , COVID-19/virología , Fármacos Cardiovasculares/uso terapéutico , Coagulación Intravascular Diseminada/diagnóstico , Coagulación Intravascular Diseminada/tratamiento farmacológico , Coagulación Intravascular Diseminada/virología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/inmunología , Hemorragia/diagnóstico , Hemorragia/tratamiento farmacológico , Hemorragia/virología , Humanos , Hiperglucemia/diagnóstico , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/virología , Inflamación , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/virología , Trombosis/diagnóstico , Trombosis/tratamiento farmacológico , Trombosis/virología , Tratamiento Farmacológico de COVID-19
16.
Heart Lung Circ ; 30(6): 786-794, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33454213

RESUMEN

The severe acute respiratory syndrome coronavirus (SARS-CoV)-2, which is responsible for coronavirus disease 2019 (COVID-19), uses angiotensin (ANG)-converting enzyme 2 (ACE2) as the entrance receptor. Although most COVID-19 cases are mild, some are severe or critical, predominantly due to acute lung injury. It has been widely accepted that a counter regulatory renin-angiotensin system (RAS) axis including the ACE2/ANG [1-7]/Mas protects the lungs from acute lung injury. However, recent evidence suggests that the generation of protective ANG [1-7] in the lungs is predominantly mediated by proinflammatory prolyl oligopeptidase (POP), which has been repeatedly demonstrated to be involved in lung pathology. This review contends that acute lung injury in severe COVID-19 is characterised by a) ACE2 downregulation and malfunction (inflammatory signalling) due to viral occupation, and b) dysregulation of the protective RAS axis, predominantly due to increased activity of proinflammatory POP. It follows that a reasonable treatment strategy in COVID-19-related acute lung injury would be delivering functional recombinant (r) ACE2 forms to trap the virus. Additionally, or alternatively to rACE2 delivery, the potential benefits resulting from lowering POP activity should also be explored. These treatment strategies deserve further investigation.


Asunto(s)
Lesión Pulmonar Aguda , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19 , Sistema Renina-Angiotensina/inmunología , Transducción de Señal , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , COVID-19/metabolismo , COVID-19/fisiopatología , COVID-19/virología , Regulación hacia Abajo , Descubrimiento de Drogas , Humanos , SARS-CoV-2/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
17.
Curr Drug Targets ; 22(3): 254-281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33081670

RESUMEN

BACKGROUND: The Coronavirus Disease 2019 (COVID-19) is becoming the major health issue in recent human history with thousands of deaths and millions of cases worldwide. Newer research and old experience with other coronaviruses highlighted a probable underlying mechanism of disturbance of the renin-angiotensin system (RAS) that is associated with the intrinsic effects of SARS-CoV-2 infection. OBJECTIVE: In this review, we aimed to describe the intimate connections between the RAS components, the immune system and COVID-19 pathophysiology. METHODS: This non-systematic review article summarizes recent evidence on the relationship between COVID-19 and the RAS. RESULTS: Several studies have indicated that the downregulation of membrane-bound ACE2 may exert a key role for the impairment of immune functions and for COVID-19 patients' outcomes. The downregulation may occur by distinct mechanisms, particularly: (1) the shedding process induced by the SARS-CoV-2 fusion pathway, which reduces the amount of membrane-bound ACE2, stimulating more shedding by the high levels of Angiotensin II; (2) the endocytosis of ACE2 receptor with the virus itself and (3) by the interferon inhibition caused by SARS-CoV-2 effects on the immune system, which leads to a reduction of ACE2 receptor expression. CONCLUSION: Recent research provides evidence of a reduction of the components of the alternative RAS axis, including ACE2 and Angiotensin-(1-7). In contrast, increased levels of Angiotensin II can activate the AT1 receptor in several organs. Consequently, increased inflammation, thrombosis and angiogenesis occur in patients infected with SARS-COV-2. Attention should be paid to the interactions of the RAS and COVID-19, mainly in the context of novel vaccines and proposed medications.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , COVID-19/inmunología , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2 , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antivirales/farmacología , COVID-19/etiología , COVID-19/genética , Regulación hacia Abajo , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Humanos , Inflamación , Ratones , Sistema Renina-Angiotensina/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Esparcimiento de Virus/efectos de los fármacos , Esparcimiento de Virus/inmunología , Tratamiento Farmacológico de COVID-19
18.
Am J Physiol Heart Circ Physiol ; 320(1): H296-H304, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33275517

RESUMEN

Biological sex is increasingly recognized as a critical determinant of health and disease, particularly relevant to the topical COVID-19 pandemic caused by the SARS-CoV-2 coronavirus. Epidemiological data and observational reports from both the original SARS epidemic and the most recent COVID-19 pandemic have a common feature: males are more likely to exhibit enhanced disease severity and mortality than females. Sex differences in cardiovascular disease and COVID-19 share mechanistic foundations, namely, the involvement of both the innate immune system and the canonical renin-angiotensin system (RAS). Immunological differences suggest that females mount a rapid and aggressive innate immune response, and the attenuated antiviral response in males may confer enhanced susceptibility to severe disease. Furthermore, the angiotensin-converting enzyme 2 (ACE2) is involved in disease pathogenesis in cardiovascular disease and COVID-19, either to serve as a protective mechanism by deactivating the RAS or as the receptor for viral entry, respectively. Loss of membrane ACE2 and a corresponding increase in plasma ACE2 are associated with worsened cardiovascular disease outcomes, a mechanism attributed to a disintegrin and metalloproteinase (ADAM17). SARS-CoV-2 infection also leads to ADAM17 activation, a positive feedback cycle that exacerbates ACE2 loss. Therefore, the relationship between cardiovascular disease and COVID-19 is critically dependent on the loss of membrane ACE2 by ADAM17-mediated proteolytic cleavage. This article explores potential mechanisms involved in COVID-19 that may contribute to sex-specific susceptibility focusing on the innate immune system and the RAS, namely, genetics and sex hormones. Finally, we highlight here the added challenges of gender in the COVID-19 pandemic.


Asunto(s)
Inmunidad Adaptativa/inmunología , Andrógenos/inmunología , Enzima Convertidora de Angiotensina 2/genética , COVID-19/inmunología , Estrógenos/inmunología , Inmunidad Innata/inmunología , Receptores de Coronavirus/genética , Proteína ADAM17/metabolismo , Inmunidad Adaptativa/genética , Andrógenos/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/genética , COVID-19/metabolismo , COVID-19/mortalidad , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Estrógenos/metabolismo , Femenino , Genes Ligados a X/genética , Genes Ligados a X/inmunología , Humanos , Inmunidad Innata/genética , Masculino , Regiones Promotoras Genéticas , Receptores de Coronavirus/metabolismo , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/inmunología , Elementos de Respuesta/genética , SARS-CoV-2/metabolismo , Índice de Severidad de la Enfermedad , Caracteres Sexuales , Factores Sexuales , Inactivación del Cromosoma X
20.
Front Immunol ; 11: 2014, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849666

RESUMEN

To date the pathophysiology of COVID-19 remains unclear: this represents a factor determining the current lack of effective treatments. In this paper, we hypothesized a complex host response to SARS-CoV-2, with the Contact System (CS) playing a pivotal role in innate immune response. CS is linked with different proteolytic defense systems operating in human vasculature: the Kallikrein-Kinin (KKS), the Coagulation/Fibrinolysis and the Renin-Angiotensin (RAS) Systems. We investigated the role of the mediators involved. CS consists of Factor XII (FXII) and plasma prekallikrein (complexed to high-molecular-weight kininogen-HK). Autoactivation of FXII by contact with SARS-CoV-2 could lead to activation of intrinsic coagulation, with fibrin formation (microthrombosis), and fibrinolysis, resulting in increased D-dimer levels. Activation of kallikrein by activated FXII leads to production of bradykinin (BK) from HK. BK binds to B2-receptors, mediating vascular permeability, vasodilation and edema. B1-receptors, binding the metabolite [des-Arg9]-BK (DABK), are up-regulated during infections and mediate lung inflammatory responses. BK could play a relevant role in COVID-19 as already described for other viral models. Angiotensin-Converting-Enzyme (ACE) 2 displays lung protective effects: it inactivates DABK and converts Angiotensin II (Ang II) into Angiotensin-(1-7) and Angiotensin I into Angiotensin-(1-9). SARS-CoV-2 binds to ACE2 for cell entry, downregulating it: an impaired DABK inactivation could lead to an enhanced activity of B1-receptors, and the accumulation of Ang II, through a negative feedback loop, may result in decreased ACE activity, with consequent increase of BK. Therapies targeting the CS, the KKS and action of BK could be effective for the treatment of COVID-19.


Asunto(s)
Betacoronavirus/metabolismo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/fisiopatología , Fibrinólisis/inmunología , Sistema Calicreína-Quinina/inmunología , Neumonía Viral/inmunología , Neumonía Viral/fisiopatología , Sistema Renina-Angiotensina/inmunología , Enzima Convertidora de Angiotensina 2 , Bradiquinina/metabolismo , COVID-19 , Permeabilidad Capilar , Proteína Inhibidora del Complemento C1 , Infecciones por Coronavirus/virología , Factor XIIa/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Quininógeno de Alto Peso Molecular/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Calicreína Plasmática/metabolismo , Neumonía Viral/virología , Precalicreína/metabolismo , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo , SARS-CoV-2 , Vasodilatación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA