Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Biol Rep ; 49(3): 2303-2309, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: covidwho-1648443

RESUMEN

Global vaccination effort and better understanding of treatment strategies provided a ray of hope for improvement in COVID-19 pandemic, however, in many countries, the disease continues to collect its death toll. The major pathogenic mechanism behind severe cases associated with high mortality is the burst of pro-inflammatory cytokines TNF, IL-6, IFNγ and others, resulting in multiple organ failure. Although the exact contribution of each cytokine is not clear, we provide an evidence that the central mediator of cytokine storm and its devastating consequences may be TNF. This cytokine is known to be involved in activated blood clotting, lung damage, insulin resistance, heart failure, and other conditions. A number of currently available pharmaceutical agents such as monoclonal antibodies and soluble TNF receptors can effectively prevent TNF from binding to its receptor(s). Other drugs are known to block NFkB, the major signal transducer molecule used in TNF signaling, or to block kinases involved in downstream activation cascades. Some of these medicines have already been selected for clinical trials, but more work is needed. A simple, rapid, and inexpensive method of directly monitoring TNF levels may be a valuable tool for a timely selection of COVID-19 patients for anti-TNF therapy.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Pandemias , SARS-CoV-2 , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Biomarcadores , COVID-19/complicaciones , COVID-19/metabolismo , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/prevención & control , Reposicionamiento de Medicamentos , Humanos , Interleucina-6/metabolismo , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Selección de Paciente , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/fisiología
2.
Lancet Diabetes Endocrinol ; 9(9): 586-594, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1545532

RESUMEN

BACKGROUND: COVID-19 can lead to multiorgan failure. Dapagliflozin, a SGLT2 inhibitor, has significant protective benefits for the heart and kidney. We aimed to see whether this agent might provide organ protection in patients with COVID-19 by affecting processes dysregulated during acute illness. METHODS: DARE-19 was a randomised, double-blind, placebo-controlled trial of patients hospitalised with COVID-19 and with at least one cardiometabolic risk factor (ie, hypertension, type 2 diabetes, atherosclerotic cardiovascular disease, heart failure, and chronic kidney disease). Patients critically ill at screening were excluded. Patients were randomly assigned 1:1 to dapagliflozin (10 mg daily orally) or matched placebo for 30 days. Dual primary outcomes were assessed in the intention-to-treat population: the outcome of prevention (time to new or worsened organ dysfunction or death), and the hierarchial composite outcome of recovery (change in clinical status by day 30). Safety outcomes, in patients who received at least one study medication dose, included serious adverse events, adverse events leading to discontinuation, and adverse events of interest. This study is registered with ClinicalTrials.gov, NCT04350593. FINDINGS: Between April 22, 2020 and Jan 1, 2021, 1250 patients were randomly assigned with 625 in each group. The primary composite outcome of prevention showed organ dysfunction or death occurred in 70 patients (11·2%) in the dapagliflozin group, and 86 (13·8%) in the placebo group (hazard ratio [HR] 0·80, 95% CI 0·58-1·10; p=0·17). For the primary outcome of recovery, 547 patients (87·5%) in the dapagliflozin group and 532 (85·1%) in the placebo group showed clinical status improvement, although this was not statistically significant (win ratio 1·09, 95% CI 0·97-1·22; p=0·14). There were 41 deaths (6·6%) in the dapagliflozin group, and 54 (8·6%) in the placebo group (HR 0·77, 95% CI 0·52-1·16). Serious adverse events were reported in 65 (10·6%) of 613 patients treated with dapagliflozin and in 82 (13·3%) of 616 patients given the placebo. INTERPRETATION: In patients with cardiometabolic risk factors who were hospitalised with COVID-19, treatment with dapagliflozin did not result in a statistically significant risk reduction in organ dysfunction or death, or improvement in clinical recovery, but was well tolerated. FUNDING: AstraZeneca.


Asunto(s)
Compuestos de Bencidrilo/administración & dosificación , COVID-19/complicaciones , Factores de Riesgo Cardiometabólico , Glucósidos/administración & dosificación , Insuficiencia Multiorgánica/prevención & control , Inhibidores del Cotransportador de Sodio-Glucosa 2/administración & dosificación , Anciano , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/complicaciones , Resultado del Tratamiento
3.
Immunopharmacol Immunotoxicol ; 43(6): 633-643, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-1467231

RESUMEN

The coronavirus disease-19 (COVID-19), at first, was reported in Wuhan, China, and then rapidly became pandemic throughout the world. Cytokine storm syndrome (CSS) in COVID-19 patients is associated with high levels of cytokines and chemokines that cause multiple organ failure, systemic inflammation, and hemodynamic instabilities. Acute respiratory distress syndrome (ARDS), a common complication of COVID-19, is a consequence of cytokine storm. In this regard, several drugs have been being investigated to suppress this inflammatory condition. Purinergic signaling receptors comprising of P1 adenosine and P2 purinoceptors play a critical role in inflammation. Therefore, activation or inhibition of some subtypes of these kinds of receptors is most likely to be beneficial to attenuate cytokine storm. This article summarizes suggested therapeutic drugs with potential anti-inflammatory effects through purinergic receptors.


Asunto(s)
Antiinflamatorios/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/sangre , Antagonistas Purinérgicos/uso terapéutico , Receptores Purinérgicos/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Animales , Antiinflamatorios/efectos adversos , Biomarcadores/sangre , COVID-19/sangre , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Interacciones Huésped-Patógeno , Humanos , Ligandos , Terapia Molecular Dirigida , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/virología , Antagonistas Purinérgicos/efectos adversos , Receptores Purinérgicos/metabolismo , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Transducción de Señal
4.
Front Immunol ; 12: 720192, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1378190

RESUMEN

COVID-19 might lead to multi-organ failure and, in some cases, to death. The COVID-19 severity is associated with a "cytokine storm." Danger-associated molecular patterns (DAMPs) are proinflammatory molecules that can activate pattern recognition receptors, such as toll-like receptors (TLRs). DAMPs and TLRs have not received much attention in COVID-19 but can explain some of the gender-, weight- and age-dependent effects. In females and males, TLRs are differentially expressed, likely contributing to higher COVID-19 severity in males. DAMPs and cytokines associated with COVID-19 mortality are elevated in obese and elderly individuals, which might explain the higher risk for severer COVID-19 in these groups. Adenosine signaling inhibits the TLR/NF-κB pathway and, through this, decreases inflammation and DAMPs' effects. As vaccines will not be effective in all susceptible individuals and as new vaccine-resistant SARS-CoV-2 mutants might develop, it remains mandatory to find means to dampen COVID-19 disease severity, especially in high-risk groups. We propose that the regulation of DAMPs via adenosine signaling enhancement might be an effective way to lower the severity of COVID-19 and prevent multiple organ failure in the absence of severe side effects.


Asunto(s)
Alarminas/inmunología , COVID-19/fisiopatología , Mediadores de Inflamación/inmunología , Adenosina/metabolismo , Alarminas/antagonistas & inhibidores , Animales , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/terapia , Humanos , Inflamación/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Gravedad del Paciente , Transducción de Señal , Receptores Toll-Like/antagonistas & inhibidores , Receptores Toll-Like/inmunología
5.
Am J Cardiovasc Drugs ; 20(6): 525-533, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-755898

RESUMEN

Human factor Xa (FXa) is a serine protease of the common coagulation pathway. FXa is known to activate prothrombin to thrombin, which eventually leads to the formation of cross-linked blood clots. While this process is important in maintaining hemostasis, excessive thrombin generation results in a host of thrombotic conditions. FXa has also been linked to inflammation via protease-activated receptors. Together, coagulopathy and inflammation have been implicated in the pathogenesis of viral infections, including the current coronavirus pandemic. Direct FXa inhibitors have been shown to possess anti-inflammatory and antiviral effects, in addition to their established anticoagulant activity. This review summarizes the pharmacological activities of direct FXa inhibitors, their pharmacokinetics, potential drug-drug interactions and adverse effects, and the details of clinical trials involving direct FXa inhibitors in coronavirus disease 2019 (COVID-19) patients.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/fisiopatología , Inhibidores del Factor Xa/farmacología , Inhibidores del Factor Xa/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/fisiopatología , Citocinas/biosíntesis , Interacciones Farmacológicas , Factor Xa/metabolismo , Inhibidores del Factor Xa/efectos adversos , Inhibidores del Factor Xa/farmacocinética , Semivida , Humanos , Mediadores de Inflamación/metabolismo , Tasa de Depuración Metabólica , Insuficiencia Multiorgánica/fisiopatología , Insuficiencia Multiorgánica/prevención & control , Pandemias , Unión Proteica/fisiología , SARS-CoV-2 , Índice de Severidad de la Enfermedad
6.
Life Sci ; 272: 119251, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1096150

RESUMEN

A novel infectious disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was detected in December 2019 and declared as a global pandemic by the World Health. Approximately 15% of patients with COVID-19 progress to severe pneumonia and eventually develop acute respiratory distress syndrome (ARDS), septic shock and/or multiple organ failure with high morbidity and mortality. Evidence points towards a determinant pathogenic role of members of the renin-angiotensin system (RAS) in mediating the susceptibility, infection, inflammatory response and parenchymal injury in lungs and other organs of COVID-19 patients. The receptor for advanced glycation end-products (RAGE), a member of the immunoglobulin superfamily, has important roles in pulmonary pathological states, including fibrosis, pneumonia and ARDS. RAGE overexpression/hyperactivation is essential to the deleterious effects of RAS in several pathological processes, including hypertension, chronic kidney and cardiovascular diseases, and diabetes, all of which are major comorbidities of SARS-CoV-2 infection. We propose RAGE as an additional molecular target in COVID-19 patients for ameliorating the multi-organ pathology induced by the virus and improving survival, also in the perspective of future infections by other coronaviruses.


Asunto(s)
COVID-19/complicaciones , Descubrimiento de Drogas , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , SARS-CoV-2/fisiología , Animales , COVID-19/metabolismo , COVID-19/patología , Humanos , Terapia Molecular Dirigida , Insuficiencia Multiorgánica/metabolismo , Insuficiencia Multiorgánica/patología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
7.
Turk J Haematol ; 38(1): 15-21, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1045314

RESUMEN

Objective: The defective interplay between coagulation and inflammation may be the leading cause of intravascular coagulation and organ dysfunction in coronavirus disease-19 (COVID-19) patients. Abnormal coagulation profiles were reported to be associated with poor outcomes. In this study, we assessed the prognostic values of antithrombin (AT) activity levels and the impact of fresh frozen plasma (FFP) treatment on outcome. Materials and Methods: Conventional coagulation parameters as well as AT activity levels and outcomes of 104 consecutive critically ill acute respiratory distress syndrome (ARDS) patients with laboratory-confirmed COVID-19 disease were retrospectively analyzed. Patients with AT activity below 75% were treated with FFP. Maximum AT activity levels achieved in those patients were recorded. Results: AT activity levels at admission were significantly lower in nonsurvivors than survivors (73% vs. 81%). The cutoff level for admission AT activity was 79% and 58% was the lowest AT for survival. The outcome in those patients who had AT activity levels above 75% after FFP treatment was better than that of the nonresponding group. As well as AT, admission values of D-dimer, C-reactive protein, and procalcitonin were coagulation and inflammatory parameters among the mortality risk factors. Conclusion: AT activity could be used as a prognostic marker for survival and organ failure in COVID-19-associated ARDS patients. AT supplementation therapy with FFP in patients with COVID-19-induced hypercoagulopathy may improve thrombosis prophylaxis and thus have an impact on survival.


Asunto(s)
Antitrombinas/sangre , COVID-19/sangre , COVID-19/terapia , Enfermedad Crítica/mortalidad , Anciano , Anciano de 80 o más Años , Antitrombinas/fisiología , Antitrombinas/uso terapéutico , Pruebas de Coagulación Sanguínea/métodos , Proteína C-Reactiva/análisis , COVID-19/diagnóstico , COVID-19/mortalidad , Estudios de Casos y Controles , Coagulación Intravascular Diseminada/etiología , Coagulación Intravascular Diseminada/prevención & control , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Plasma , Polipéptido alfa Relacionado con Calcitonina/análisis , Pronóstico , Estudios Retrospectivos , SARS-CoV-2/genética , Trombofilia/complicaciones , Trombofilia/fisiopatología , Turquia/epidemiología
8.
Cytokine Growth Factor Rev ; 58: 114-133, 2021 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1007960

RESUMEN

The devastating global impact of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has prompted scientists to develop novel strategies to fight Coronavirus Disease of 2019 (COVID-19), including the examination of pre-existing treatments for other viral infections in COVID-19 patients. This review provides a reasoned discussion of the possible use of Mesenchymal Stromal Cells (MSC) or their products as a treatment in SARS-CoV-2-infected patients. The main benefits and concerns of using this cellular therapy, guided by preclinical and clinical data obtained from similar pathologies will be reviewed. MSC represent a highly immunomodulatory cell population and their use may be safe according to clinical studies developed in other pathologies. Notably, four clinical trials and four case reports that have already been performed in COVID-19 patients obtained promising results. The clinical application of MSC in COVID-19 is very preliminary and further investigational studies are required to determine the efficacy of the MSC therapy. Nevertheless, these preliminary studies were important to understand the therapeutic potential of MSC in COVID-19. Based on these encouraging results, the United States Food and Drug Administration (FDA) authorized the compassionate use of MSC, but only in patients with Acute Respiratory Distress Syndrome (ARDS) and a poor prognosis. In fact, patients with severe SARS-CoV-2 can present infection and tissue damage in different organs, such as lung, heart, liver, kidney, gut and brain, affecting their function. MSC may have pleiotropic activities in COVID-19, with the capacity to fight inflammation and repair lesions in several organs.


Asunto(s)
COVID-19/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , COVID-19/epidemiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Trasplante de Células Madre Mesenquimatosas/tendencias , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/terapia , SARS-CoV-2/patogenicidad
9.
Med Hypotheses ; 146: 110455, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: covidwho-969218

RESUMEN

SARS-CoV-2 infection generally begins in the respiratory tract where it can cause bilateral pneumonia. The disease can evolve into acute respiratory distress syndrome and multi-organ failure, due to viral spread in the blood and an excessive inflammatory reaction including cytokine storm. Antiviral and anti-cytokine drugs have proven to be poorly or in-effective in stopping disease progression, and mortality or serious chronic damage is common in severely ill cases. The low efficacy of antiviral drugs is probably due to late administration, when the virus has triggered the inflammatory reaction and is no longer the main protagonist. The relatively poor efficacy of anti-cytokine drugs is explained by the fact that they act on one or a few of the dozens of cytokines involved, and because other mediators of inflammation - reactive oxygen and nitrogen species - are not targeted. When produced in excess, reactive species cause extensive cell and tissue damage. The only drug known to inhibit the excessive production of reactive species and cytokines is methylene blue, a low-cost dye with antiseptic properties used effectively to treat malaria, urinary tract infections, septic shock, and methaemoglobinaemia. We propose testing methylene blue to contrast Covid-related acute respiratory distress syndrome, but particularly suggest testing it early in Covid infections to prevent the hyper-inflammatory reaction responsible for the serious complications of the disease.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Azul de Metileno/farmacología , Modelos Biológicos , Antivirales/farmacología , COVID-19/complicaciones , COVID-19/fisiopatología , Citocinas/antagonistas & inhibidores , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/lesiones , Humanos , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Pandemias , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2 , Insuficiencia del Tratamiento
10.
Biomaterials ; 267: 120389, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-898508

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new strain of coronavirus not previously identified in humans. Globally, the number of confirmed cases and mortality rates of coronavirus disease 2019 (COVID-19) have risen dramatically. Currently, there are no FDA-approved antiviral drugs and there is an urgency to develop treatment strategies that can effectively suppress SARS-CoV-2-mediated cytokine storms, acute respiratory distress syndrome (ARDS), and sepsis. As symptoms progress in patients with SARS-CoV-2 sepsis, elevated amounts of cell-free DNA (cfDNA) are produced, which in turn induce multiple organ failure in these patients. Furthermore, plasma levels of DNase-1 are markedly reduced in SARS-CoV-2 sepsis patients. In this study, we generated recombinant DNase-1-coated polydopamine-poly(ethylene glycol) nanoparticulates (named long-acting DNase-1), and hypothesized that exogenous administration of long-acting DNase-1 may suppress SARS-CoV-2-mediated neutrophil activities and the cytokine storm. Our findings suggest that exogenously administered long-acting nanoparticulate DNase-1 can effectively reduce cfDNA levels and neutrophil activities and may be used as a potential therapeutic intervention for life-threatening SARS-CoV-2-mediated illnesses.


Asunto(s)
COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , ADN/sangre , Desoxirribonucleasa I/uso terapéutico , Portadores de Fármacos/administración & dosificación , Nanopartículas/administración & dosificación , Neutrófilos/efectos de los fármacos , SARS-CoV-2 , Sepsis/tratamiento farmacológico , Animales , COVID-19/sangre , COVID-19/inmunología , Síndrome de Liberación de Citoquinas/etiología , Desoxirribonucleasa I/administración & dosificación , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Trampas Extracelulares/efectos de los fármacos , Humanos , Indoles , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , FN-kappa B/sangre , Neutrófilos/enzimología , Peroxidasa/sangre , Polietilenglicoles , Poliglactina 910 , Polímeros , Sepsis/etiología , Sepsis/inmunología
11.
J Clin Pharm Ther ; 46(2): 440-446, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: covidwho-887386

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: The coronavirus disease 2019 (COVID-19) associated cytokine activation can lead to a rapid progression into respiratory failure, shock and multiorgan failure. Interleukin-6 (IL-6) is a pro-inflammatory cytokine that likely contributes to the pathogenesis of cytokine release syndrome. It is hypothesized that modulating IL-6 levels or its effects with tocilizumab, a recombinant humanized anti-IL-6 receptor monoclonal antibody, may alter the course of disease. METHODS: We examined the association between tocilizumab use and intubation or death at a community hospital in New York City. Data were obtained regarding consecutive patients hospitalized with COVID-19. The primary end point was a composite of intubation or death in a time-to-event analysis. We compared outcomes in patients who received tocilizumab with those in patients who did not, using a multivariable Cox model with inverse probability weighting according to the propensity score. RESULTS AND DISCUSSION: In this single-centre retrospective cohort study involving 1225 hospitalized patients with SARS-CoV-2 infection, the probability to respiratory failure, which was measured as intubation or death, was less frequent in patients who received tocilizumab. WHAT IS NEW AND CONCLUSION: Tocilizumab and other IL-6 receptor monoclonal antibodies may evolve as a viable option in treating patients with moderate and severe COVID-19.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Tratamiento Farmacológico de COVID-19 , COVID-19 , Síndrome de Liberación de Citoquinas , Interleucina-6 , Respiración Artificial , SARS-CoV-2 , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/mortalidad , Correlación de Datos , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/etiología , Femenino , Mortalidad Hospitalaria , Humanos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/efectos adversos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Ciudad de Nueva York/epidemiología , Respiración Artificial/métodos , Respiración Artificial/estadística & datos numéricos , Estudios Retrospectivos , SARS-CoV-2/aislamiento & purificación , Resultado del Tratamiento
13.
Life Sci ; 256: 117907, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: covidwho-626534

RESUMEN

Acute lung injury (ALI) and the subsequent multi-system organ failure is a serious health problem with devastating impacts on the health care systems. Indeed, the world has been facing an un-preceded situation in the past couple of months following COVID-19 infestation and the associated high-mortality rates mainly attributed to sepsis and the associated multiple organ failures of particular concern; acute respiratory distress syndrome post lung injury. The current study provides evidence on the ameliorative impact of nifuroxazide, and FDA approved antidiarrheal drug in attenuation of lipopolysaccharide (LPS)-induced ALI and myocarditis when administrated either in prophylactic or curative regimens. Nifuroxazide administration was associated with a significant improvement in lung and heart histopathological characteristics and architecture with retraction of LPS-induced inflammatory-infiltration. This was associated with retraction in serum biomarkers of cellular injury of which; LDH, CK-MB, and ALP. Nifuroxazide administration was associated with a significant improvement in both lung and heart oxidative status. Such positive outcomes were underlined by a significant inhibitory effect of nifuroxazide on lung and heart contents of toll-like receptor (4) (TLR4)/the inflammasome NALPR3/interleukin- 1ß (IL-1ß). In conclusion: Nifuroxazide attenuates LPS-induced ALI and myocardial injury via interruption of TLR4/NALPR3/IL-1ß signaling. Thus it can offer a potential approach for attenuation of sepsis in critically ill patients.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Infecciones por Coronavirus/complicaciones , Hidroxibenzoatos/farmacología , Miocarditis/prevención & control , Nitrofuranos/farmacología , Neumonía Viral/complicaciones , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Animales , COVID-19 , Infecciones por Coronavirus/epidemiología , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Miocarditis/etiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pandemias , Neumonía Viral/epidemiología , Ratas , Ratas Sprague-Dawley , Sepsis/complicaciones , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
14.
Protein Cell ; 11(10): 707-722, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-626150

RESUMEN

The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/complicaciones , Trasplante de Células Madre Mesenquimatosas , Pandemias , Neumonía Viral/complicaciones , Síndrome de Dificultad Respiratoria/terapia , Traslado Adoptivo , Células Epiteliales Alveolares/patología , Animales , Apoptosis , Líquidos Corporales/metabolismo , Linfocitos T CD4-Positivos/inmunología , COVID-19 , Ensayos Clínicos como Asunto , Coinfección/prevención & control , Coinfección/terapia , Infecciones por Coronavirus/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Oxigenación por Membrana Extracorpórea , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Neumonía Viral/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/patología , SARS-CoV-2 , Investigación Biomédica Traslacional
15.
Nitric Oxide ; 102: 39-41, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: covidwho-600932

RESUMEN

COVID-19 is a severe pandemic which has caused a devastating amount of loss in lives around the world, and yet we still don't know how to appropriately treat this disease. We know very little about the pathogenesis of SARS-CoV-2, the virus which induces the COVID-19. However, COVID-19 does share many similar symptoms with SARS and influenza. Previous scientific discoveries learned from lab animal models and clinical practices shed light on possible pathogenic mechanisms in COVID-19. In the past decades, accumulated scientific findings confirmed the pathogenic role of free radicals damage in respiratory virus infection. Astonishingly very few medical professionals mention the crucial role of free radical damage in COVID-19. This hypothesis aims to summarize the crucial pathogenic role of free radical damage in respiratory virus induced pneumonia and suggest an antioxidative therapeutic strategy for COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/metabolismo , Radicales Libres/metabolismo , Pandemias , Neumonía Viral/metabolismo , Acetilcisteína/uso terapéutico , Animales , Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Azitromicina/administración & dosificación , Azitromicina/farmacología , Azitromicina/uso terapéutico , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/metabolismo , Quimioterapia Combinada , Radicales Libres/antagonistas & inhibidores , Glutatión/uso terapéutico , Humanos , Hidroxicloroquina/administración & dosificación , Hidroxicloroquina/farmacología , Hidroxicloroquina/uso terapéutico , Ratones , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Factor 2 Relacionado con NF-E2/agonistas , Óxido Nítrico/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Estrés Oxidativo , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/metabolismo , Tratamiento Farmacológico de COVID-19
16.
Expert Opin Ther Targets ; 24(8): 723-730, 2020 08.
Artículo en Inglés | MEDLINE | ID: covidwho-620479

RESUMEN

INTRODUCTION: The cytokine release syndrome (CRS) of COVID-19 is associated with the development of critical illness requiring multi-organ support. Further research is required to halt progression of multi-organ injury induced by hyper-inflammation. AREAS COVERED: PubMed/MEDLINETM databases were accessed between May 9th-June 9th, 2020, to review the latest perspectives on the treatment and pathogenesis of CRS. EXPERT OPINION: Over-activity of chemotaxis triggers a macrophage activation syndrome (MAS) resulting in the release of pro-inflammatory cytokines. IL-6 and TNF- α are at the forefront of hyper-inflammation. The inflammatory cascade induces endothelial activation and capillary leak, leading to circulatory collapse and shock. As endothelial dysfunction persists, there is activation of the clotting cascade and microvascular obstruction. Continued endothelial activation results in multi-organ failure, regardless of pulmonary tissue damage. We propose that targeting the endothelium may interrupt this cycle. Immuno-modulating therapies have been suggested, however, further data is necessary to confirm that they do not jeopardize adaptive immunity. Inhibition of IL-6 and the Janus Kinase, signal transducer and activator of transcription proteins pathway (JAK/STAT), are favorable targets. Remote ischemic conditioning (RIC) reduces the inflammation of sepsis in animal models and should be considered as a low risk intervention, in combination with cardiovascular protection.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Síndrome de Liberación de Citoquinas , Insuficiencia Multiorgánica , Pandemias , Manejo de Atención al Paciente/métodos , Neumonía Viral , Animales , Betacoronavirus/patogenicidad , Betacoronavirus/fisiología , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/virología , Neumonía Viral/inmunología , Neumonía Viral/terapia , Neumonía Viral/virología , SARS-CoV-2 , Transducción de Señal/inmunología
17.
Naunyn Schmiedebergs Arch Pharmacol ; 393(7): 1157-1160, 2020 07.
Artículo en Inglés | MEDLINE | ID: covidwho-361504

RESUMEN

Vitamin D is an immunomodulator hormone with an anti-inflammatory and antimicrobial effect with a high safety profile. A lot of COVID-19 infected patients develop acute respiratory distress syndrome (ARDS), which may lead to multiple organ damage. These symptoms are associated with a cytokine storm syndrome. The aim of this letter is to note the 5 crucial points that vitamin D could have protective and therapeutic effects against COVID-19. For that reason, COVID-19 infection-induced multiple organ damage might be prevented by vitamin D.


Asunto(s)
Antiinflamatorios/administración & dosificación , Infecciones por Coronavirus/complicaciones , Neumonía Viral/complicaciones , Vitamina D/administración & dosificación , Antiinflamatorios/farmacología , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/prevención & control , Síndrome de Liberación de Citoquinas/virología , Humanos , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/virología , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Síndrome de Dificultad Respiratoria/prevención & control , Síndrome de Dificultad Respiratoria/virología , Vitamina D/farmacología , Tratamiento Farmacológico de COVID-19
18.
Biomed Pharmacother ; 127: 110195, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: covidwho-154554

RESUMEN

Since early December 2019, a number of pneumonia cases associated with unknown coronavirus infection were identified in Wuhan, China, and many additional cases were identified in other regions of China and in other countries within 3 months. Currently, more than 80,000 cases have been diagnosed in China, including more than 3000 deaths. The epidemic is spreading to the rest of the world, posing a grave challenge to prevention and control. On February 12, 2020, the International Committee on Taxonomy of Viruses and the World Health Organization officially named the novel coronavirus and associated pneumonia as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease 2019 (COVID-19), respectively. According to the recent research on SARS-CoV-2, the virus mainly infects the respiratory system but may cause damage to other systems. In this paper, we will systematically review the pathogenic features, transmission routes, and infection mechanisms of SARS-CoV-2, as well as any adverse effects on the digestive system, urogenital system, central nervous system, and circulatory system, in order to provide a theoretical and clinical basis for the diagnosis, classification, treatment, and prognosis assessment of SARS-CoV-2 infection.


Asunto(s)
Betacoronavirus , Sistema Cardiovascular/virología , Sistema Nervioso Central/virología , Infecciones por Coronavirus , Sistema Digestivo/virología , Insuficiencia Multiorgánica , Pandemias , Neumonía Viral , Sistema Urogenital/virología , Betacoronavirus/aislamiento & purificación , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/transmisión , Manejo de la Enfermedad , Humanos , Insuficiencia Multiorgánica/prevención & control , Insuficiencia Multiorgánica/virología , Neumonía Viral/epidemiología , Neumonía Viral/fisiopatología , Neumonía Viral/terapia , Neumonía Viral/transmisión , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA