Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Chinese Journal of Biotechnology ; (12): 2669-2683, 2023.
Article in Chinese | WPRIM | ID: wpr-981224

ABSTRACT

The goal of this study was to investigate the regulatory effect of angiotensin converting enzyme 2 (ACE2) on cellular inflammation caused by avian infectious bronchitis virus (IBV) and the underlying mechanism of such effect. Vero and DF-1 cells were used as test target to be exposed to recombinant IBV virus (IBV-3ab-Luc). Four different groups were tested: the control group, the infection group[IBV-3ab-Luc, MOI (multiplicity of infection)=1], the ACE2 overexpression group[IBV-3ab Luc+pcDNA3.1(+)-ACE2], and the ACE2-depleted group (IBV-3ab-Luc+siRNA-ACE2). After the cells in the infection group started to show cytopathic indicators, the overall protein and RNA in cell of each group were extracted. real-time quantitative polymerase chain reaction (RT-qPCR) was used to determine the mRNA expression level of the IBV nucleoprotein (IBV-N), glycoprotein 130 (gp130) and cellular interleukin-6 (IL-6). Enzyme linked immunosorbent assay (ELISA) was used to determine the level of IL-6 in cell supernatant. Western blotting was performed to determine the level of ACE2 phosphorylation of janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). We found that ACE2 was successfully overexpressed and depleted in both Vero and DF-1 cells. Secondly, cytopathic indicators were observed in infected Vero cells including rounding, detaching, clumping, and formation of syncytia. These indicators were alleviated in ACE2 overexpression group but exacerbated when ACE2 was depleted. Thirdly, in the infection group, capering with the control group, the expression level of IBV-N, gp130, IL-6 mRNA and increased significantly (P < 0.05), the IL-6 level was significant or extremely significant elevated in cell supernatant (P < 0.05 or P < 0.01); the expression of ACE2 decreased significantly (P < 0.05); protein phosphorylation level of JAK2 and STAT3 increased significantly (P < 0.05). Fourthly, comparing with the infected group, the level of IBV-N mRNA expression in the ACE2 overexpression group had no notable change (P > 0.05), but the expression of gp130 mRNA, IL-6 level and expression of mRNA were elevated (P < 0.05) and the protein phosphorylation level of JAK2 and STAT3 decreased significantly (P < 0.05). In the ACE2-depleted group, there was no notable change in IBV-N (P > 0.05), but the IL-6 level and expression of mRNA increased significantly (P < 0.05) and the phosphorylation level of JAK2 and STAT3 protein decreased slightly (P > 0.05). The results demonstrated for the first time that ACE2 did not affect the replication of IBV in DF-1 cell, but it did contribute to the prevention of the activation of the IL-6/JAK2/STAT3 signaling pathway, resulting in an alleviation of IBV-induced cellular inflammation in Vero and DF-1 cells.


Subject(s)
Animals , Humans , Chlorocebus aethiops , Interleukin-6/genetics , Janus Kinase 2/pharmacology , Infectious bronchitis virus/metabolism , STAT3 Transcription Factor/metabolism , Angiotensin-Converting Enzyme 2/pharmacology , Cytokine Receptor gp130/metabolism , Vero Cells , Signal Transduction , Inflammation , RNA, Messenger
2.
Chinese Critical Care Medicine ; (12): 665-668, 2023.
Article in Chinese | WPRIM | ID: wpr-982651

ABSTRACT

Coronaviruses are single-stranded RNA viruses that are common in animals. In the past 20 years, there have been three large-scale epidemics of coronaviruses, including severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and coronavirus disease (COVID). Heart disease is an independent risk factor for severe COVID. At the same time, SARS-CoV-2 infection is often complicated with myocardial injury, which is closely related to poor prognosis. The receptors of SARS coronavirus are angiotensin-converting enzyme 2 (ACE2) and CD209L, among which ACE2 is the main receptor, and ACE2 is abundant in the heart. The receptor of MERS-coronavirus is dipeptide peptidase 4 (DPP4), which is not expressed in myocardial cells, but existed in vascular endothelial cells and blood. These receptors are important factors for the myocardial injury caused by coronavirus infection.


Subject(s)
Animals , COVID-19 , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , Endothelial Cells , Peptidyl-Dipeptidase A/genetics
3.
Rev. Hosp. Ital. B. Aires (2004) ; 42(4): 231-239, dic. 2022. ilus, tab
Article in Spanish | LILACS, UNISALUD, BINACIS | ID: biblio-1424871

ABSTRACT

INTRODUCCIÓN: Un nuevo brote de coronavirus surgió en 2019 en Wuhan, China, causando conmoción en el sistema sanitario de todo el mundo; el Comité Internacional de Taxonomía de Virus lo denominó SARS-CoV-2, agente causante de la enfermedad COVID-19.El espectro de gravedad de la enfermedad es muy amplio: la mayoría de los pacientes no presentan gravedad, pero otros pueden desarrollar neumonías, y la insuficiencia respiratoria aguda es la causa más frecuente de mortalidad. Objetivo: analizar y desarrollar las distintas alternativas terapéuticas aportadas por la Biotecnología para tratar los síntomas de aquellos pacientes con COVID-19. Metodología: se realizó una revisión de la bibliografía disponible, a partir de enero de 2020 en PubMed, acerca de los tratamientos que se encuentran aún en ensayos clínicos y aquellos que cuentan con aprobación bajo uso de emergencia para la enfermedad COVID-19. También se realizaron búsquedas a través de Google y Google Académico para publicaciones de organismos de Salud en referencia a políticas de salud establecidas para la terapéutica durante dicha pandemia. Resultados: este trabajo aborda las nuevas alternativas terapéuticas para COVID-19 derivadas de la Biotecnología, que se encuentran tanto en uso como en etapas de ensayos clínicos comprendidos dentro del segmento de los biofármacos y las bioterapias. Se incluye un breve resumen del estatus regulatorio de entidades de salud, el mecanismo de acción de dichas terapias y características generales de cada uno. Se incluyen novedosas bioterapias que se empezaron a implementar para afrontar la pandemia. Conclusiones: la pandemia de coronavirus está poniendo a prueba el sistema sanitario internacional, para brindar soluciones tanto desde el diagnóstico y prevención como para el tratamiento de la población a fin de disminuir la mortalidad. Esto incluyó, obviamente también, al área de la Biotecnología aplicada a la salud, que ha aportado en los tres aspectos mencionados; el presente trabajo se centra en las respuestas de tipo terapéutico que ha brindado y que están comercializadas o en fases clínicas. (AU)


INTRODUCTION: A new coronavirus outbreak emerged in 2019 in Wuhan, China, causing a shock to the healthcare system around the world; the International Committee on Taxonomy of Viruses named it SARS-CoV- 2, the infectious agent of the COVID-19 disease. The spectrum of severity of the disease is very wide, most patients are not serious, but others can develop pneumonia, with acute respiratory failure being the most frequent cause of mortality. Objective: to analyze and develop the different therapeutic alternatives provided by Biotechnology dedicated to Health, to treat the symptoms of those COVID-19 patients who require it, and thus reduce mortality.Methodology: a review of the available literature from January 2020 in PubMed of the treatments that are still in clinical trials and those that have been approved under emergency use for the disease COVID-19 was performed. Searches were also carried out through Google and Google Scholar for publications of Health organizations in reference to health policies established for therapeutics during the mentioned pandemic. Results: this work addresses the new therapeutic alternatives derived from Biotechnology, which are both in use and in stages of clinical trials, to treat patients who developed COVID-19 included within the segment of biopharmaceuticals and biotherapies. A brief summary of the regulatory status of health entities, the mechanism of action of said therapies and general characteristics of each one is included. Innovative biotherapies that began to be implemented to face the pandemic are included. Conclusions: The coronavirus pandemic has driven the international health system to the test, to provide solutions both from the diagnosis, prevention and treatment of the population to reduce the mortality of patients. This obviously also included the area of Biotechnology applied to health, which has contributed in the three aspects mentioned. The present work focuses on the therapeutic responses that it has provided and that are commercialized or in clinical phases. (AU)


Subject(s)
Humans , Animals , Biological Products/therapeutic use , Biological Therapy/methods , Adrenal Cortex Hormones/therapeutic use , SARS-CoV-2/drug effects , COVID-19/drug therapy , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Biological Therapy/classification , Biological Therapy/standards , Biotechnology , Clinical Trials as Topic , Peptidyl-Dipeptidase A/drug effects , Angiotensin-Converting Enzyme 2/drug effects , Immunomodulating Agents/therapeutic use , COVID-19 Serotherapy , Horses , Immune Sera/biosynthesis , Antibodies, Monoclonal/therapeutic use
4.
Journal of Southern Medical University ; (12): 860-867, 2022.
Article in Chinese | WPRIM | ID: wpr-941014

ABSTRACT

OBJECTIVE@#To investigate the roles of angiotensin-converting enzyme 2 (ACE2) in ozone-induced pulmonary inflammation and airway remodeling in mice.@*METHODS@#Sixteen wild-type (WT) C57BL/6J mice and 16 ACE2 knock-out (KO) mice were exposed to either filtered air or ozone (0.8 ppm) for 3 h per day for 5 consecutive days. Masson's staining and HE staining were used to observe lung pathologies. Bronchoalveolar lavage fluid (BALF) was collected and the total cell count was determined. The total proteins and cytokines in BALF were determined by BCA and ELISA method. The transcription levels of airway remodeling-related indicators in the lung tissues were detected using real-time quantitative PCR. The airway resistance of the mice was measured using a small animal ventilator with methacholine stimulation.@*RESULTS@#Following ozoneexposure ACE2 KO mice had significantly higher lung pathological scores than WT mice (P < 0.05). Masson staining results showed that compared with ozone-exposed WT mice, ozone-exposed ACE2 KO mice presented with significantly larger area of collagen deposition in the bronchi [(19.62±3.16)% vs (6.49±1.34)%, P < 0.05] and alveoli [(21.63±3.78)% vs (4.44±0.99)%, P < 0.05]. The total cell count and total protein contents in the BALF were both higher in ozone-exposed ACE2 KO mice than in WT mice, but these differences were not statistically significant (P > 0.05). The concentrations of IL-6, IL-1β, TNF-α, CXCL1/KC and MCP-1 in the BALF were all higher in ozone-exposed ACE2 KO mice than in ozone-exposed WT mice, but only the difference in IL-1β was statistically significant (P < 0.05). The transcription levels of MMP-9, MMP-13, TIMP 4, COL1A1, and TGF-β in the lung tissues were all significantly higher in ozone-exposed ACE2 KO mice (P < 0.01). No significant difference was found in airway resistance between ozone-exposed ACE KO mice and WT mice after challenge with 0, 10, 25, or 100 mg/mL of methacholine.@*CONCLUSION@#ACE2 participates in ozone-induced lung inflammation and airway remodeling in mice.


Subject(s)
Animals , Mice , Airway Remodeling , Angiotensin-Converting Enzyme 2 , Methacholine Chloride , Mice, Inbred C57BL , Mice, Knockout , Ozone/adverse effects , Pneumonia
5.
J. bras. nefrol ; 43(3): 303-310, July-Sept. 2021. tab, graf
Article in English, Portuguese | LILACS | ID: biblio-1340129

ABSTRACT

Abstract Introduction: Sickle cell nephropathy begins in childhood and presents early increases in glomerular filtration, which, over the long term, can lead to chronic renal failure. Several diseases have increased circulating and urinary angiotensin-converting enzyme (ACE) activity, but there is little information about changes in ACEs activity in children with sickle cell disease (SCD). Objective: We examined circulating and urinary ACE 1 activity in children with SCD. Methods: This cross-sectional study compared children who were carriers of SCD with children who comprised a control group (CG). Serum and urinary activities of ACE were evaluated, as were biochemical factors, urinary album/creatinine rates, and estimated glomerular filtration rate. Results: Urinary ACE activity was significantly higher in patients with SCD than in healthy children (median 0.01; range 0.00-0.07 vs median 0.00; range 0.00-0.01 mU/mL·creatinine, p < 0.001. No significant difference in serum ACE activities between the SCD and CG groups was observed (median 32.25; range 16.2-59.3 vs median 40.9; range 18.0-53.4) mU/m`L·creatinine, p < 0.05. Conclusion: Our data revealed a high urinary ACE 1 activity, different than plasmatic level, in SCD patients suggesting a dissociation between the intrarenal and systemic RAAS. The increase of urinary ACE 1 activity in SCD patients suggests higher levels of Ang II with a predominance of classical RAAS axis, that can induce kidney damage.


Resumo Introdução: A nefropatia falciforme começa na infância e apresenta aumentos precoces na filtração glomerular, que, em longo prazo, podem levar à insuficiência renal crônica. Várias doenças têm aumentado a atividade da enzima conversora da angiotensina (ECA) urinária e circulante, mas há pouca informação sobre alterações na atividade das ECAs em crianças com doença falciforme (DF). Objetivo: Examinamos a atividade da ECA-1 circulante e urinária em crianças com DF. Métodos: Este estudo transversal comparou crianças que eram portadoras de DF com crianças que compunham um Grupo Controle (GC). As atividades séricas e urinárias da ECA foram avaliadas, assim como os fatores bioquímicos, a relação albumina/creatinina urinária e a taxa de filtração glomerular estimada. Resultados: A atividade urinária da ECA foi significativamente maior em pacientes com DF do que em crianças saudáveis (mediana 0,01; intervalo 0,00-0,07 vs mediana 0,00; intervalo 0,00-0,01 mU/mL·creatinina, p < 0,001. Não foi observada diferença significativa nas atividades séricas da ECA entre os grupos DF e GC (mediana 32,25; intervalo 16,2-59,3 vs mediana 40,9; intervalo 18,0-53,4) mU/mL·creatinina, p < 0,05. Conclusão: Nossos dados revelaram uma alta atividade urinária da ECA-1, diferente do nível plasmático, em pacientes com DF, sugerindo uma dissociação entre o Sistema Renina Angiotensina Aldosterona (SRAA) intra-renal e sistêmico. O aumento da atividade urinária da ECA-1 em pacientes com DF sugere níveis mais elevados de Ang II com predominância do eixo clássico do SRAA, que pode induzir lesão renal.


Subject(s)
Humans , Child , Renal Insufficiency, Chronic , Anemia, Sickle Cell , Angiotensins , Cross-Sectional Studies , Peptidyl-Dipeptidase A , Angiotensin-Converting Enzyme 2
6.
J. bras. nefrol ; 43(2): 254-262, Apr.-June 2021. tab
Article in English | LILACS | ID: biblio-1286931

ABSTRACT

ABSTRACT Covid-19 has been identified as the cause of acute respiratory disease with interstitial and alveolar pneumonia, but it can affect several organs, such as kidneys, heart, blood, nervous system and digestive tract. The disease-causing agent (Sars-CoV-2) has a binding structure to the angiotensin-converting enzyme 2 (ACE2) receptor, enabling entry into cells that express ACE2, such as the pulmonary alveolar epithelial cells. However, studies also indicate the possibility of damage to renal cells, since these cells express high levels of ACE2. Currently, there is no evidence to indicate a specific treatment for covid-19. Several drugs have been used, and some of them may have their excretion process altered in patients with abnormal kidney function. To date, there are no studies that assist health professionals in adjusting the dose of these drugs. Thus, this study aims to review and discuss the topic, taking into account factors associated with kidney injury in covid-19, as well as pharmacokinetic aspects and dose recommendations of the main drugs used for covid-19.


RESUMO A covid-19 foi identificada como a causa de doença respiratória aguda com pneumonia intersticial e alveolar, mas que pode afetar vários órgãos, como rim, coração, sangue, sistema nervoso e trato digestivo. O agente causador da doença (Sars-CoV-2) tem uma estrutura de ligação ao receptor da enzima de conversão da angiotensina 2 (ACE2), permitindo a entrada em células que expressam ACE2, como as células epiteliais alveolares pulmonares. Porém, estudos também indicam a possibilidade de lesão das células renais, uma vez que essas células expressam altos níveis de ACE2. Atualmente, não existem evidências para a indicação de um tratamento específico para a covid-19. Vários medicamentos vêm sendo utilizados, e alguns podem ter o processo de eliminação alterados em pacientes com comprometimento renal. Até o momento, não há estudos que auxiliem os profissionais de saúde no ajuste de dose desses medicamentos. Assim, este estudo tem como objetivo revisar e discutir o tema, levando em consideração os fatores relacionados à lesão renal na covid-19, bem como aspectos farmacocinéticos e recomendações de doses dos principais medicamentos utilizados para covid-19.


Subject(s)
Humans , COVID-19/complications , COVID-19/drug therapy , Kidney/physiopathology , Kidney Diseases/virology , Angiotensin-Converting Enzyme 2
7.
Rev. méd. Chile ; 149(5): 733-737, mayo 2021. tab
Article in Spanish | LILACS | ID: biblio-1389510

ABSTRACT

Background: Angiotensin-converting enzyme 2 (ACE2) has a protective role in the regulation of blood pressure. Aim: To evaluate the influence of ACE2 rs1978124 gene polymorphism on the hemodynamic response after a six-minute walk submaximal aerobic test. Material and Methods: A six-minute walk submaximal aerobic test was carried out in 47 men and 55 women, aged 22 ± 2 years. Blood pressure was recorded before and after the test. Pulse rate was recorded continuously. ACE2 polymorphism was determined in DNA extracted from a blood sample. Results: For women, the genotype distribution did not deviate from Hardy-Weinberg equilibrium (x2 = 0.804, d.f. = 1, P = 0.4) and no significant differences in heart rate, systolic and diastolic blood pressure were observed between CC, CT and TT genotypes before and after the test. Among men no differences in these parameters were observed either between the three phenotypes. Conclusions: ACE2 rs1978124 gene polymorphism did not influence the hemodynamic response to submaximal exercise in these participants.


Subject(s)
Humans , Male , Female , Young Adult , Exercise , Angiotensin-Converting Enzyme 2/genetics , Polymorphism, Genetic , Blood Pressure , Genotype , Hemodynamics
8.
Bol. méd. Hosp. Infant. Méx ; 78(2): 91-94, Mar.-Apr. 2021.
Article in English | LILACS | ID: biblio-1249112

ABSTRACT

Abstract Coronavirus disease 2019 (COVID-19) is a new disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. China reported the first case of COVID-19 in December 2019, and a few months later, the World Health Organization declared it as a pandemic. Oral ulcers in adult patients have been associated with COVID-19. However, no cases have yet been documented in children. The angiotensin-converting enzyme-2 (ACE2) receptor has been identified in tissues of the oral cavity. Studies have identified the tongue as the site with the highest expression of ACE2, and the oral epithelium, gingival epithelium, and salivary glands as sites of lesser extent expression. ACE2 expression is lower in children and varies with age. SARS-CoV-2 in saliva has been identified in various studies, which suggests that this could be a useful sample for diagnosis. However, its presence in saliva would indicate the high risk of contagion of this fluid.


Resumen La COVID-19 es una nueva enfermedad causada por el SARS-CoV-2 (coronavirus tipo 2 del síndrome respiratorio agudo grave). El primer caso de COVID-19 se reportó en China en diciembre de 2019, y unos meses después la Organización Mundial de la Salud la declaró como una pandemia. En pacientes adultos se han asociado úlceras orales a la COVID-19; en niños aún no se han documentado casos. El receptor de la enzima convertidora de la angiotensina 2 (ECA2) se ha identificado en tejidos de la cavidad oral. Los estudios han identificado que la lengua es el sitio con mayor expresión del receptor de la ECA2, y el epitelio bucal, el epitelio gingival y las glándulas salivales lo son en menor medida. La expresión de la ECA2 es menor en los niños y va aumentando con la edad. En diversos estudios se ha identificado el SARS-CoV-2 en la saliva, lo que sugiere que podría ser una muestra útil para el diagnóstico de este virus. Sin embargo, su presencia en saliva indicaría un alto riesgo de contagio de este fluido.


Subject(s)
Adult , Child , Humans , Oral Health , Oral Ulcer/virology , SARS-CoV-2/isolation & purification , COVID-19/complications , Saliva/virology , Age Factors , Angiotensin-Converting Enzyme 2/metabolism , COVID-19 Testing , COVID-19/diagnosis , COVID-19/virology , Mouth/virology
9.
Arq. Asma, Alerg. Imunol ; 5(1): 66-78, jan.mar.2021. ilus
Article in Portuguese | LILACS | ID: biblio-1398415

ABSTRACT

No combate à infecção pelo coronavírus 2 da síndrome respiratória aguda grave (SARS-CoV-2), o organismo se utiliza de mecanismos da imunidade inata, dentre eles os receptores Toll- Like (TLR), responsáveis pela sinalização da inflamação através da liberação de mediadores químicos e recrutamento de células imunitárias. Na patologia causada pela doença do SARS-CoV-2 2019 (COVID-19), ganha especial importância o TLR-4, visto que a sua estimulação exacerbada vem sendo relacionada ao estado hiperinflamatório em fases avançadas da COVID-19. Outro receptor que desempenha um papel primordial na infecção pelo SARS-CoV-2, servindo como porta de entrada para o vírus e progressão da doença, é a enzima conversora de angiotensina 2 (ECA 2), cuja ligação com a proteína S viral causa desregulação de vários sistemas fundamentais para a homeostase, como o sistema renina-angiotensina-aldosterona. Pacientes com doenças cardiometabólicas como obesidade, diabetes, aterosclerose e hipertensão vêm sendo classificados como alto risco para desenvolver as formas graves da COVID-19, visto que o estado inflamatório, já existente nessas doenças, pode ser agravado pelo desequilíbrio metabólico causado pelo SARS-CoV-2. A elucidação desses e de outros mecanismos relacionados à fisiopatologia da COVID-19 é imprescindível para uma melhora na estratificação de risco, nas escolhas terapêuticas e no prognóstico desses pacientes. Desta forma, nesta revisão objetivamos discutir as relações entre TLR-4, ECA 2, doenças cardiometabólicas, infecção pelo SARS-CoV-2 e gravidade da COVID-19.


In the fight against the infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the body uses mechanisms from the innate immune system, such as Toll-Like receptors (TLR), responsible for inflammation signaling through release of chemical mediators and recruitment of immune cells. In the disease caused by SARS-CoV-2 (COVID-19), TLR-4 assumes special importance because its exacerbated stimulation has been related to a hyperinflammatory state in advanced stages of COVID-19. Another receptor that plays a major role in SARS-CoV-2 infection, serving as a gateway to the virus and impacting disease progression, is angiotensin-converting enzyme 2 (ACE-2), whose binding to the viral S protein causes dysregulation of several key systems for homeostasis, such as the renin-angiotensin-aldosterone system. The elucidation of these and other mechanisms related to the pathophysiology of COVID-19 is essential for an improvement in risk stratification, therapeutic choices, and prognosis for these patients. Thus, we aimed to discuss in this review the relationships between TLR-4, ACE-2, cardiometabolic diseases, SARS-CoV-2 infection, and severity of COVID-19.


Subject(s)
Humans , Diabetes Mellitus , Atherosclerosis , Toll-Like Receptors , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , COVID-19 , COVID-19/physiopathology , Hypertension , Obesity , Pathology , Patients , Prognosis , Renin-Angiotensin System , Therapeutics , Viruses , Immune System
10.
Rev. cuba. med ; 60(supl.1): e2484, 2021. graf
Article in Spanish | LILACS, CUMED | ID: biblio-1408965

ABSTRACT

Introducción: La pandemia derivada de la enfermedad por el nuevo coronavirus 2019 (COVID-19) se ha convertido en una emergencia de salud pública mundial, debido a que puede desarrollar complicaciones que amenazan la vida. Si bien se sabe que el SARS-CoV-2 causa enfermedad pulmonar sustancial, se han observado muchas manifestaciones extrapulmonares, incluyendo el compromiso del sistema gastrointestinal. El megacolon tóxico es una complicación rara pero, potencialmente, mortal que se asocia más con la enfermedad inflamatoria intestinal. Sin embargo, cualquier afección que conduzca a la inflamación del colon puede conducir a una dilatación tóxica. Objetivo: Se presenta el caso de un paciente con un síndrome de dificultad respiratoria aguda secundario a una infección por SARS-COV-2. De manera concomitante presentó un cuadro de dilatación no obstructiva del colon, asociado con toxicidad sistémica. Caso clínico: El desarrollo de megacolon tóxico en un paciente con SARS-COV-2 puede estar justificado debido a que el virus infecta las células huésped a través del receptor de la enzima convertidora de angiotensina 2. Se cumplieron los criterios diagnósticos para megacolon tóxico. Conclusiones: Esta también se encuentra altamente expresada en las células epiteliales intestinales, por lo tanto, se debe considerar su diagnóstico oportuno para una intervención temprana, en aras de reducir la tasa de mortalidad tanto como sea posible(AU)


Introduction: The pandemic derived from the 2019 novel coronavirus disease (COVID-19) has become a global public health emergency, due to the fact that it can develop life-threatening complications. Although SARS-CoV-2 is known to cause substantial lung disease, many extra-pulmonary manifestations have been observed, including involvement of the gastrointestinal system. Toxic mega colon is a rare but life-threatening complication most associated with inflammatory bowel disease. However, any condition that leads to inflammation of the colon can lead to toxic dilation. Objective: To report the case of a patient with ARDS secondary to a SARS-COV-2 infection. Concomitantly, she had non-obstructive dilation of the colon, associated with systemic toxicity. Clinical case report: The development of toxic mega colon in a patient with SARS-COV-2 may be justified because the virus infects host cells through the angiotensin-converting enzyme 2 receptor. The diagnostic criteria for toxic megacolon were met. Conclusions: It is also highly expressed in intestinal epithelial cells, therefore, its timely diagnosis should be considered for early intervention, in order to reduce the mortality rate as much as possible(AU)


Subject(s)
Humans , Gastrointestinal Diseases/epidemiology , Angiotensin-Converting Enzyme 2 , COVID-19/complications , Megacolon, Toxic/epidemiology , Ecuador
11.
Rev inf cient ; 100(5): 1-12, 2021. ilus
Article in Spanish | LILACS, CUMED | ID: biblio-1348804

ABSTRACT

Introducción: La COVID-19 causada por el virus del SARS-CoV-2 es una pandemia que ha cobrado la vida de millones de personas y sobrecargado los servicios sanitarios de todo el mundo. Objetivo: Describir la relación entre la proteína de la espícula (proteína S, proteína espicular o spike) del SARS-CoV-2 y enzima convertidora de angiotensina 2 como desencadenante primario de la infección por la COVID-19. Método: Se realizó una búsqueda bibliográfica en Google Académico, SciELO y PubMed, con los descriptores iniciales COVID-19 y SARS-CoV-2. El periodo de publicación seleccionado fue entre los años 2019-2021, sin restricciones en cuanto al tipo de artículo. Los trabajos debieron estar disponibles en español e inglés a texto completo. Resultados: La proteína de la espícula del SARS-CoV-2, que desempeña un papel clave en el reconocimiento del receptor y en el proceso de fusión de la membrana celular, está compuesta por dos subunidades, S1 y S2. La subunidad S1 contiene un dominio de unión al receptor RBD (por sus siglas en inglés, receptor-binding domain) que se une al receptor del huésped, la enzima convertidora de angiotensina 2, mientras que la subunidad S2 interviene en la fusión de la membrana viral y celular. La ubicuidad tisular de la enzima convertidora de angiotensina 2 explica las múltiples manifestaciones clínicas de la enfermedad. Conclusiones: El conocimiento de la relación entre el SARS-CoV-2 y su receptor enzima convertidora de angiotensina 2 permite no solo conocer la fisiopatología de la COVID-19, sino el diseño de fármacos antivirales y vacunas que contribuyen a la prevención y tratamiento de esta enfermedad viral.


Introduction: COVID-19 caused by the SARS-CoV-2 virus is a pandemic that has claimed the lives of millions of people and overloaded health services around the world. Objective: To describe the relationship between the spike protein (S) of SARS-CoV-2 and the angiotensin-converting enzyme 2 as the primary trigger of COVID-19 infection. Method: A bibliographic search was carried out in Google Scholar, SciELO and PubMed, with the initial descriptors COVID-19 and SARS-CoV-2. The publication period selected was between the years 2019 to 2021, without restrictions regarding the type of article. The papers had to be available in full text in Spanish and English. Results: The spike protein of SARS-CoV-2, which plays a key role in receptor recognition and in the cell membrane fusion process, is composed of two subunits, S1 and S2. The S1 subunit contains a receptor-binding domain (RBD) that binds to the host's receptor, angiotensin-converting enzyme 2, while the S2 subunit is involved in the viral and cellular membrane fusion. The tissue ubiquity of angiotensin converting enzyme 2 explains the multiple clinical manifestations of the disease. Conclusions: The knowledge of the relationship between SARS-CoV-2 and its receptor the angiotensin-converting enzyme 2, allows not only to know the pathophysiology of COVID-19, but also the design of antiviral drugs and vaccines that contribute to the prevention and treatment of this viral disease.


Introdução: COVID-19 causada pelo vírus SARS-CoV-2 é uma pandemia que ceifou a vida de milhões de pessoas e sobrecarregou os serviços de saúde em todo o mundo. Objetivo: Descrever a relação entre a proteína spike (S) do SARS-CoV-2 e a enzima conversora de angiotensina 2 como o principal fator desencadeante da infecção por COVID-19. Método: Foi realizada uma busca bibliográfica no Google Scholar, SciELO e PubMed, com os descritores iniciais COVID-19 e SARS-CoV-2. O período de publicação selecionado foi entre os anos de 2019 a 2021, sem restrições quanto ao tipo de artigo. Os artigos deveriam estar disponíveis na íntegra em espanhol e inglês. Resultados: A proteína spike do SARS-CoV-2, que desempenha um papel fundamental no reconhecimento do receptor e no processo de fusão da membrana celular, é composta por duas subunidades, S1 e S2. A subunidade S1 contém um domínio de ligação ao receptor (RBD) que se liga ao receptor do hospedeiro, a enzima conversora de angiotensina 2, enquanto a subunidade S2 está envolvida na fusão da membrana viral e celular. A onipresença tecidual da enzima conversora da angiotensina 2 explica as múltiplas manifestações clínicas da doença. Conclusões: O conhecimento da relação entre o SARS-CoV-2 e seu receptor, a enzima conversora de angiotensina 2, permite não só conhecer a fisiopatologia da COVID-19, mas também o desenho de antivirais e vacinas que contribuam para a prevenção e tratamento desta doença viral.


Subject(s)
Spike Glycoprotein, Coronavirus/analysis , Spike Glycoprotein, Coronavirus/classification , Spike Glycoprotein, Coronavirus/ultrastructure , Angiotensin-Converting Enzyme 2/physiology , COVID-19 Vaccines , COVID-19/physiopathology
12.
Afr. J. Clin. Exp. Microbiol ; 22(4): 423-429, 2021.
Article in English | AIM | ID: biblio-1342104

ABSTRACT

Severe acute respiratory syndrome­coronavirus-2 (SARS-CoV-2) enters cells using the angiotensin converting enzyme 2 (ACE2), which are expressed by the respiratory tract endothelium, epithelial cells of the stomach, duodenum, ileum, rectum, cholangiocytes, and hepatocytes. Pathological examinations of these organs are not feasible method of diagnosis but can explain pathological changes, pathogenesis of the disease, and the cause of death in COVID-19 cases. In this review, we performed a literature search for COVID-19-related pathological changes seen during post-mortem examinations in different organs of the body including the lungs, gastrointestinal tract, liver, kidney, skin, heart and blood. Our findings showed that SARS-CoV-2 has damaging effects on many organs, probably due to the host immune responses to the presence of the virus. It is recommended that both antiviral and immunomodulatory agents should be considered in the management of COVID-19 patients for better prognosis, and clinical outcome


Subject(s)
Humans , Pathology , Angiotensin-Converting Enzyme 2 , COVID-19 , Patients , Autopsy , Nigeria
13.
Afr. J. Clin. Exp. Microbiol ; 22(4): 430-438, 2021.
Article in English | AIM | ID: biblio-1342105

ABSTRACT

Because of high mortality and long-term hospital stay among patients with SARS-CoV-2 infections, it is important to search for biochemical changes in different organs and systems that could be useful in diagnosis and prognosis of COVID-19. We conducted a literature search of online databases including PubMed, Web of Science, Scopus and Google scholar for relevant materials on biochemical changes in SARS-COV-2 infections published between December 2019 and March 2021. The review shows that SARS-COV-2 uses the angiotensin converting enzyme 2 (ACE2) for attachment and entry into host cells. These ACE2 are abundantly expressed by the epithelial cells of the respiratory tract and moderately expressed by the epithelial cells of the esophagus, stomach, duodenum, ileum, rectum, cholangiocytes, liver hepatocytes, pancreatic beta cells, and kidney tubular cells. This explains the systemic nature of SARS-COV-2 infection, and the high morbidity and mortality associated with COVID-19. Although, tests to assess biochemical changes are not specific enough for the diagnosis of SARS-CoV-2 infection, they may be useful for predicting outcome of COVID-19. This review highlights biochemical parameters that are significantly elevated or reduced in SARS-COV-2 infections, and which can be used as predictive factors of the severity and prognosis in COVID-19 patients.


Subject(s)
Humans , Biomarkers , Angiotensin-Converting Enzyme 2 , COVID-19 , Diagnosis
14.
Chinese Medical Journal ; (24): 935-943, 2021.
Article in English | WPRIM | ID: wpr-878142

ABSTRACT

BACKGROUND@#Since 2019, a novel coronavirus named 2019 novel coronavirus (2019-nCoV) has emerged worldwide. Apart from fever and respiratory complications, acute kidney injury has been observed in a few patients with coronavirus disease 2019. Furthermore, according to recent findings, the virus has been detected in urine. Angiotensin-converting enzyme II (ACE2) has been proposed to serve as the receptor for the entry of 2019-nCoV, which is the same as that for the severe acute respiratory syndrome. This study aimed to investigate the possible cause of kidney damage and the potential route of 2019-nCoV infection in the urinary system.@*METHODS@#We used both published kidney and bladder cell atlas data and new independent kidney single-cell RNA sequencing data generated in-house to evaluate ACE2 gene expression in all cell types in healthy kidneys and bladders. The Pearson correlation coefficients between ACE2 and all other genes were first generated. Then, genes with r values larger than 0.1 and P values smaller than 0.01 were deemed significant co-expression genes with ACE2.@*RESULTS@#Our results showed the enriched expression of ACE2 in all subtypes of proximal tubule (PT) cells of the kidney. ACE2 expression was found in 5.12%, 5.80%, and 14.38% of the proximal convoluted tubule cells, PT cells, and proximal straight tubule cells, respectively, in three published kidney cell atlas datasets. In addition, ACE2 expression was also confirmed in 12.05%, 6.80%, and 10.20% of cells of the proximal convoluted tubule, PT, and proximal straight tubule, respectively, in our own two healthy kidney samples. For the analysis of public data from three bladder samples, ACE2 expression was low but detectable in bladder epithelial cells. Only 0.25% and 1.28% of intermediate cells and umbrella cells, respectively, had ACE2 expression.@*CONCLUSION@#This study has provided bioinformatics evidence of the potential route of 2019-nCoV infection in the urinary system.


Subject(s)
Humans , Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , Gene Expression , Kidney/metabolism , SARS-CoV-2 , Sequence Analysis, RNA , Single-Cell Analysis , Urinary Bladder/metabolism
15.
Journal of Zhejiang University. Science. B ; (12): 330-340, 2021.
Article in English | WPRIM | ID: wpr-880733

ABSTRACT

Epidemiological evidence suggests that patients with hypertension infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are at increased risk of acute lung injury. However, it is still not clear whether this increased risk is related to the usage of renin-angiotensin system (RAS) blockers. We collected medical records of coronavirus disease 2019 (COVID-19) patients from the First Affiliated Hospital, Zhejiang University School of Medicine (Hangzhou, China), and evaluated the potential impact of an angiotensin II receptor blocker (ARB) on the clinical outcomes of COVID-19 patients with hypertension. A total of 30 hypertensive COVID-19 patients were enrolled, of which 17 were classified as non-ARB group and the remaining 13 as ARB group based on the antihypertensive therapies they received. Compared with the non-ARB group, patients in the ARB group had a lower proportion of severe cases and intensive care unit (ICU) admission as well as shortened length of hospital stay, and manifested favorable results in most of the laboratory testing. Viral loads in the ARB group were lower than those in the non-ARB group throughout the disease course. No significant difference in the time of seroconversion or antibody levels was observed between the two groups. The median levels of soluble angiotensin-converting enzyme 2 (sACE2) in serum and urine samples were similar in both groups, and there were no significant correlations between serum sACE2 and biomarkers of disease severity. Transcriptional analysis showed 125 differentially expressed genes which mainly were enriched in oxygen transport, bicarbonate transport, and blood coagulation. Our results suggest that ARB usage is not associated with aggravation of COVID-19. These findings support the maintenance of ARB treatment in hypertensive patients diagnosed with COVID-19.


Subject(s)
Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme 2/blood , Antibodies, Viral/blood , Antihypertensive Agents/therapeutic use , Biomarkers , COVID-19/complications , China , Hypertension/drug therapy , Intensive Care Units , Length of Stay , Retrospective Studies , Transcriptome , Viral Load
17.
Gac. méd. Méx ; 156(4): 324-329, Jul.-Aug. 2020. graf
Article in English | LILACS | ID: biblio-1249919

ABSTRACT

Abstract In the efforts to explain COVID-19 pathophysiology, studies are being carried out on the correspondence between the expression of SARS-CoV-2 cell receptors and viral sequences. ACE2, CD147 and TMPRSS2 receptors expression could indicate poorly explored potential infection targets. For the genomic analysis of SARS-CoV-2 receptors, using BioGPS information was decided, which is a portal that centralizes genetic annotation resources, in combination with that of The Human Protein Atlas, the largest portal of human transcriptome and proteome data. We also reviewed the most recent articles on the subject. RNA and viral receptor proteins expression was observed in numerous anatomical sites, which partially coincides with the information reported in the literature. High expression in testicular cells markedly stood out, and it would be therefore important ruling out whether this anatomical site is a SARS-CoV-2 reservoir; otherwise, germ cell damage, as it is observed in infections with other RNA viruses, should be determined.


Resumen En el afán por explicar la fisiopatogenia de COVID-19 se están realizando estudios en torno a la correspondencia entre la expresión de receptores celulares de SARS-CoV-2 y las secuencias virales. La expresión de los receptores ACE2, CD147 y TMPRSS2 podría indicar blancos de infección poco explorados. Para el análisis genómico de los receptores de SARS-CoV-2 se optó por utilizar la información del BioGPS, un portal que centraliza los recursos de anotación genética, en combinación con la de The Human Protein Atlas, el portal más grande de datos del transcriptoma y proteoma humanos. También se revisaron los artículos más recientemente respecto al tema. En numerosos sitios anatómicos se observó la expresión de ARN y proteínas de los receptores del virus, que coinciden parcialmente con la información reportada en la literatura. Resaltó la alta expresión en las células de los testículos, por lo que sería importante descartar si este sitio anatómico es un reservorio de SARS-CoV-2; de no ser así, determinar el daño en las células germinales, tal como sucede en infecciones por otros virus ARN.


Subject(s)
Humans , Pneumonia, Viral/virology , Testis/virology , Coronavirus Infections/virology , Betacoronavirus/isolation & purification , Pneumonia, Viral/physiopathology , Serine Endopeptidases/genetics , Gene Expression Regulation , Virus Latency , Coronavirus Infections/physiopathology , Peptidyl-Dipeptidase A/genetics , Basigin/genetics , Pandemics , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , COVID-19
18.
Gac. méd. Méx ; 156(4): 348-351, Jul.-Aug. 2020. graf
Article in English | LILACS | ID: biblio-1249923

ABSTRACT

Abstract Introduction: Reports of dermatological manifestations in patients with COVID-19 suggest a possible cutaneous tropism of SARS-CoV-2; however, the capacity of this virus to infect the skin is unknown. Objective: To determine the susceptibility of the skin to SARS-CoV-2 infection based on the expression of viral entry factors ACE2 and TMPRSS2 in this organ. Method: A comprehensive analysis of human tissue gene expression databases was carried out looking for the presence of the ACE2 and TMPRSS2 genes in the skin. mRNA expression of these genes in skin-derived human cell lines was also assessed. Results: The analyses showed high co-expression of ACE2 and TMPRSS2 in the gastrointestinal tract and kidney, but not in the skin. Only the human immortalized keratinocyte HaCaT cell line expressed detectable levels of ACE2, and no cell line originating in the skin expressed TMPRSS2. Conclusions: Our results suggest that cutaneous manifestations in patients with COVID-19 cannot be directly attributed to the virus. It is possible that cutaneous blood vessels endothelial damage, as well as the effect of circulating inflammatory mediators produced in response to the virus, are the cause of skin involvement.


Resumen Introducción: Reportes de manifestaciones dermatológicas en pacientes con COVID-19 sugieren un posible tropismo cutáneo del virus SARS-CoV-2; sin embargo, se desconoce la capacidad de este virus para infectar la piel. Objetivo: Determinar la susceptibilidad de la piel a la infección por SARS-CoV-2 con base en la expresión de los factores de entrada viral ACE2 y TMPRSS2 en dicho órgano. Método: Se buscaron los genes ACE2 y TMPRSS2 en la piel, para lo cual se realizó un análisis extenso de las bases de datos de expresión genética en tejidos humanos. Asimismo, se evaluó la expresión de dichos genes en líneas celulares humanas derivadas de la piel. Resultados: Los análisis mostraron alta expresión conjunta de ACE2 y TMPRSS2 en el tracto gastrointestinal y en los riñones, pero no en la piel. Solo la línea celular de queratinocitos humanos inmortalizados HaCaT expresó niveles detectables de ACE2 y ninguna línea celular de origen cutáneo expresó TMPRSS2. Conclusiones: Los resultados sugieren que las manifestaciones dermatológicas en pacientes con COVID-19 no pueden ser atribuidas directamente al virus; es posible que sean originadas por el daño endotelial a los vasos sanguíneos cutáneos y el efecto de los mediadores inflamatorios circulantes producidos en respuesta al virus.


Subject(s)
Humans , Pneumonia, Viral/complications , Serine Endopeptidases/genetics , Skin Diseases, Viral/virology , Coronavirus Infections/complications , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/genetics , Skin/virology , Cell Line , Gene Expression Regulation , Coronavirus Infections/genetics , Genetic Predisposition to Disease , Virus Internalization , Viral Tropism/physiology , Pandemics , Betacoronavirus/isolation & purification , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , COVID-19
19.
Rev. chil. pediatr ; 91(3): 330-338, jun. 2020. graf
Article in Spanish | LILACS | ID: biblio-1126169

ABSTRACT

Resumen: El sistema renina angiotensina aldosterona (SRAA) es el principal regulador del volumen plasmático, manteniendo la homeostasis cardiovascular e hidrosalina. En la vía clásica, la enzima convertidora de angiotensina (ECA) genera Angiotensina II (AngII), de potente efecto inflamatorio y vasoconstrictor. Esta vía clásica es a su vez regulada por la ECA2, que convierte AngII a Ang 1-7, cuyas acciones vaso dilatadoras y antiinflamatorias dan balance a los efectos de AngII. La ECA2 se ha relacionado con la patogenia de infecciones respiratorias como el virus respiratorio sincicial y el síndrome respiratorio agudo grave por coronavirus (SARS-CoV y SARS-CoV-2). Estudios recientes han demostrado que la ECA2 corresponde al principal receptor del SARS-CoV-2, que en conjunto con otros receptores como la serin proteasa TMPRSS2, permiten la fijación, fusión y entrada del virus a la célula huésped. En animales infectados por SARS-CoV se produce una caída de la concentración tisular de ECA2 y Ang 1-7, con la consiguiente sobreexpresión de AngII, y sus efectos vasoconstrictores e inflamatorios. Experimentos con ECA2 recombinante han mostrado un efecto protector frente a la sobreexpresión del SRAA en animales infectados por SARS-CoV, efecto similar al demostrado con el uso de bloquea- dores del receptor de AngII, AT1. La evidencia sobre el rol protector de ECA2 parece respaldar las recomendaciones respecto a no suspender estos medicamentos en la infección SARS-CoV-2. En este artículo presentamos el conocimiento actual sobre el rol del SRAA en la infección por SARS-CoV, a partir de conceptos fisiopatológicos, bases moleculares, y evidencia experimental y clínica.


Abstract: The renin-angiotensin-aldosterone system (RAAS) is the main plasma volume regulator, which maintains cardiovascular and hydrosaline homeostasis. In the classical pathway, the angiotensin converting enzyme (ACE) generates Angiotensin II (AngII), which is powerfully inflammatory and vasoconstrictive. This classical pathway is also regulated by ACE2, which converts AngI to Ang 1-9, and degrades AngII to Ang 1-7, whose vasodilatory and anti-inflammatory functions balance out the effects of AngII. ACE2 has been associated with the pathogenesis of respiratory infections such as RSV and severe acute respiratory syndrome coronavirus (SARS-CoV and SARS-CoV-2). Recent studies have shown that ACE2 corresponds to the main SARS-CoV-2 receptor, which together with other receptors such as the TMPRSS2, allows the virus to attach, fuse, and enter the host cell. These studies have shown that in animals infected with coronavirus there is a drop in tissue concentration of ACE2 and Ang 1-7, leading to overexpression of AngII and its vasoconstrictive and inflammatory effects. Experiments with recombinant ACE2 have shown a protective effect against overexpression of RAAS in coronavirus-infected animals, which is similar to that demonstrated with the use of AnglI receptor blockers (AT1). Evidence on the protective role of ACE2 seems to support the recommendations re garding not discontinuing these drugs in COVID-19 infection. In this article, we present the current knowledge about the role of RAAS in coronavirus infection, based on physiopathological concepts, molecular bases, and experimental and clinical evidence.


Subject(s)
Humans , Animals , Pneumonia, Viral/virology , Coronavirus Infections/virology , Peptidyl-Dipeptidase A/metabolism , Betacoronavirus/isolation & purification , Pneumonia, Viral/physiopathology , Renin-Angiotensin System/physiology , Coronavirus Infections/physiopathology , Angiotensin Receptor Antagonists/pharmacology , Pandemics , Angiotensin-Converting Enzyme 2 , SARS-CoV-2 , COVID-19
SELECTION OF CITATIONS
SEARCH DETAIL