Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Exp Biol Med (Maywood) ; 248(4): 309-316, 2023 02.
Artículo en Inglés | MEDLINE | ID: covidwho-2229355

RESUMEN

Heme-oxygenase 1 (HO-1) is an enzyme with well-known anti-inflammatory and antioxidant properties, whose levels have been previously associated with disease severity in the context of sterile and infectious diseases. Moreover, the heme/HO-1 pathway has been associated with prothrombotic changes in other diseases. Accordingly, the potential of modulating HO-1 levels for the treatment of COVID-19 was extensively speculated during the COVID-19 pandemic, but very few actual data were generated. The aim of our study was to explore the association of HO-1, heme, and hemopexin (HPX) levels with COVID-19 severity and with markers of inflammation and coagulation activation. The study was conducted in 30 consecutive patients with COVID-19 admitted due to hypoxemia, and 30 healthy volunteers matched by sex, age, and geographic region. HO-1 and HPX levels were measured by enzyme immunoassay (ELISA) and heme levels were measured by a colorimetric method. A comprehensive panel of coagulation and fibrinolysis activation was also used. Patients with COVID-19 presented increased levels of HO-1 when compared to controls (5741 ± 2696 vs 1953 ± 612 pg/mL, respectively, P < 0.0001), as well as a trend toward increased levels of HPX (3.724 ± 0.880 vs 3.254 ± 1.022 mg/mL, respectively; P = 0.06). In addition, HO-1 and HPX levels reduced from admission to day + 4. HO-1 levels were associated with duration of intensive care unit stay and with several markers of coagulation activation. In conclusion, modulation of HO-1 could be associated with the prothrombotic state observed in COVID-19, and HO-1 could also represent a relevant biomarker for COVID-19. New independent studies are warranted to explore and expand these findings.


Asunto(s)
COVID-19 , Hemo , Humanos , Biomarcadores , Hemopexina/metabolismo , Pandemias , Gravedad del Paciente , Hemo-Oxigenasa 1/metabolismo
2.
Adv Exp Med Biol ; 1395: 111-116, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2173626

RESUMEN

COVID-19 is a pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The persistent and excessive inflammatory response can build up a clinical picture that is difficult to manage and potentially fatal. Potent activators of inflammatory phenomena are damage-associated molecular patterns (DAMPs) and, in particular, the high-mobility group box 1 (HMGB1). HMGB1 is an intranuclear protein that is either passively released during hypoxia-related necrosis or actively released by macrophages. Heme oxygenase (HO-1) has an anti-inflammatory effect by inhibiting HMGB1, which could be a therapeutic target to reduce COVID-19 inflammation. In our study, we evaluated CD3, CD4, CD8, HMGB1 and HO-1 in the COVID-19 lung and correlated it to clinical data.


Asunto(s)
COVID-19 , Proteína HMGB1 , Síndrome de Dificultad Respiratoria , Humanos , COVID-19/complicaciones , SARS-CoV-2/metabolismo , Hemo-Oxigenasa 1/metabolismo
3.
PLoS One ; 17(8): e0273500, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2002334

RESUMEN

BACKGROUND AND OBJECTIVE: A cytokine storm is caused by inflammatory cells, including pro-inflammatory macrophage phenotype (M1), and play a critical role in the pathogenesis of COVID-19, in which diffuse alveolar damage occurs in the lungs due to oxidative stress exposure. Heme oxygenase (HO)-1 is a stress-induced protein produced by the anti-inflammatory / anti-oxidative macrophage phenotype (M2), which also produces soluble CD163 (sCD163). In our study, we investigated and determined that serum HO-1 can be a predictive biomarker for assessing both the severity and the outcome of COVID-19 patients. METHOD: The serum concentrations of HO-1 and sCD163 of COVID-19 patients were measured on admission. The relationship between these biomarkers and other clinical parameters and outcomes were evaluated. RESULTS: Sixty-four COVID-19 patients (11 mild, 38 moderate, and 15 severe cases) were assessed. The serum HO-1 tended to increase (11.0 ng/mL vs. 24.3 ng/mL vs. 59.6 ng/mL with severity). Serum HO-1 correlated with serum lactate dehydrogenase (R = 0.422), C-reactive protein (R = 0.463), and the ground glass opacity (GGO) and consolidation score (R = 0.625) of chest computed tomography. The serum HO-1 showed a better area under the curve (AUC) for predicting ICU admission than the serum sCD163 (HO-1; 0.816 and sCD163; 0.743). In addition, composite parameters including serum HO-1 and the GGO and consolidation score showed a higher AUC for predicting ICU admission than the AUC of a single parameter. CONCLUSION: Clinically, serum HO-1, reflecting the activation of M2, could be a very useful marker for evaluating disease severity and predicting prognoses for COVID-19 patients. In addition, controlling activated M2 might be a preventative COVID-19 therapeutic target.


Asunto(s)
COVID-19 , Hemo-Oxigenasa 1/metabolismo , Biomarcadores , Humanos , Macrófagos/metabolismo , Pronóstico
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(3): 166322, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1637812

RESUMEN

BACKGROUND: Acute kidney injury (AKI) is both a consequence and determinant of outcomes in COVID-19. The kidney is one of the major organs infected by the causative virus, SARS-CoV-2. Viral entry into cells requires the viral spike protein, and both the virus and its spike protein appear in the urine of COVID-19 patients with AKI. We examined the effects of transfecting the viral spike protein of SARS-CoV-2 in kidney cell lines. METHODS: HEK293, HEK293-ACE2+ (stably overexpressing ACE2), and Vero E6 cells having endogenous ACE2 were transfected with SARS-CoV-2 spike or control plasmid. Assessment of gene and protein expression, and syncytia formation was performed, and the effects of quercetin on syncytia formation examined. FINDINGS: Spike transfection in HEK293-ACE2+ cells caused syncytia formation, cellular sloughing, and focal denudation of the cell monolayer; transfection in Vero E6 cells also caused syncytia formation. Spike expression upregulated potentially nephrotoxic genes (TNF-α, MCP-1, and ICAM1). Spike upregulated the cytoprotective gene HO-1 and relevant signaling pathways (p-Akt, p-STAT3, and p-p38). Quercetin, an HO-1 inducer, reduced syncytia formation and spike protein expression. INTERPRETATION: The major conclusions of the study are: 1) Spike protein expression in kidney cells provides a relevant model for the study of maladaptive and adaptive responses germane to AKI in COVID-19; 2) such spike protein expression upregulates HO-1; and 3) quercetin, an HO-1 inducer, may provide a clinically relevant/feasible protective strategy in AKI occurring in the setting of COVID-19. FUNDING: R01-DK119167 (KAN), R01-AI100911 (JPG), P30-DK079337; R01-DK059600 (AA).


Asunto(s)
COVID-19/metabolismo , Hemo-Oxigenasa 1/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Células Vero , Internalización del Virus/efectos de los fármacos
5.
Expert Rev Anti Infect Ther ; 20(5): 657-661, 2022 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1585374

RESUMEN

INTRODUCTION: : 5-Aminolevulinic acid (5-ALA) is a naturally synthesized amino acid present in most plants as well as animals, and it is routinely consumed by humans. This brief report sought to describe the potential of 5-ALA and sodium-ferrous citrate (5-ALA/SFC) to ameliorate the course of coronavirus disease 2019 (COVID-19). AREAS COVERED: : Studies have shown that 5-ALA is converted to protoporphyrin IX (PPIX), then to heme. Recent studies have demonstrated that PPIX has antiviral effects against several viruses, including Zika virus, dengue virus, and influenza A virus. The anti-inflammatory effects of 5-ALA have also been reported in humans. Preliminary in vitro and clinical studies have shown that the combination of 5-ALA/SFC could reduce severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-mediated insults. The SARS-CoV-2 genome contains guanine-quadruplex sequences, and the administration of 5-ALA/SFC can lead to the generation of porphyrins that have the ability to bind to guanine-quadruplexes and reduce the replication of SARS-CoV-2. Furthermore, 5-ALA is a metabolic precursor of heme, which is a potent inducer of the enzyme heme oxygenase-1, the levels of which are decreased in patients with severe COVID-19. Oral administration of 5-ALA/SFC induced heme oxygenase-1 in the peripheral blood of uninfected healthy individuals. EXPERT OPINION: : Based on the available information, it appears likely that 5-ALA/SFC has therapeutic value in clinically controlling SARS-CoV-2-mediated insults in COVID-19 patients. Multicenter randomized controlled trials are needed for determining the long-term clinical utility of 5-ALA/SFC.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Infección por el Virus Zika , Virus Zika , Ácido Aminolevulínico/farmacología , Animales , Ácido Cítrico , Compuestos Ferrosos , Hemo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , SARS-CoV-2 , Sodio
6.
Sci Rep ; 11(1): 21462, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1500517

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease-19 (COVID-19). More than 143 million cases of COVID-19 have been reported to date, with the global death rate at 2.13%. Currently, there are no licensed therapeutics for controlling SARS-CoV-2 infection. The antiviral effects of heme oxygenase-1 (HO-1), a cytoprotective enzyme that inhibits the inflammatory response and reduces oxidative stress, have been investigated in several viral infections. To confirm whether HO-1 suppresses SARS-CoV-2 infection, we assessed the antiviral activity of hemin, an effective and safe HO-1 inducer, in SARS-CoV-2 infection. We found that treatment with hemin efficiently suppressed SARS-CoV-2 replication (selectivity index: 249.7012). Besides, the transient expression of HO-1 using an expression vector also suppressed the growth of the virus in cells. Free iron and biliverdin, which are metabolic byproducts of heme catalysis by HO-1, also suppressed the viral infection. Additionally, hemin indirectly increased the expression of interferon-stimulated proteins known to restrict SARS-CoV-2 replication. Overall, the findings suggested that HO-1, induced by hemin, effectively suppressed SARS-CoV-2 in vitro. Therefore, HO-1 could be potential therapeutic candidate for COVID-19.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Hemo-Oxigenasa 1/metabolismo , Hemina/uso terapéutico , Animales , Antivirales/química , Antivirales/farmacología , COVID-19/virología , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Hemina/química , Hemina/farmacología , Humanos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Viral/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Regulación hacia Arriba/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
7.
Int J Mol Sci ; 22(4)2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1094246

RESUMEN

Since Yachie et al. reported the first description of human heme oxygenase (HO)-1 deficiency more than 20 years ago, few additional human cases have been reported in the literature. A detailed analysis of the first human case of HO-1 deficiency revealed that HO-1 is involved in the protection of multiple tissues and organs from oxidative stress and excessive inflammatory reactions, through the release of multiple molecules with anti-oxidative stress and anti-inflammatory functions. HO-1 production is induced in vivo within selected cell types, including renal tubular epithelium, hepatic Kupffer cells, vascular endothelium, and monocytes/macrophages, suggesting that HO-1 plays critical roles in these cells. In vivo and in vitro studies have indicated that impaired HO-1 production results in progressive monocyte dysfunction, unregulated macrophage activation and endothelial cell dysfunction, leading to catastrophic systemic inflammatory response syndrome. Data from reported human cases of HO-1 deficiency and numerous studies using animal models suggest that HO-1 plays critical roles in various clinical settings involving excessive oxidative stress and inflammation. In this regard, therapy to induce HO-1 production by pharmacological intervention represents a promising novel strategy to control inflammatory diseases.


Asunto(s)
Anemia Hemolítica/metabolismo , Trastornos del Crecimiento/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Estrés Oxidativo , Animales , Humanos , Inflamación
8.
Biomed Pharmacother ; 137: 111384, 2021 May.
Artículo en Inglés | MEDLINE | ID: covidwho-1082719

RESUMEN

Antiviral agents with different mechanisms of action could induce synergistic effects against SARS-CoV-2 infection. Some reports suggest the therapeutic potential of the heme oxygenase-1 (HO-1) enzyme against virus infection. Given that hemin is a natural inducer of the HO-1 gene, the aim of this study was to develop an in vitro assay to analyze the antiviral potency of hemin against SARS-CoV-2 infection. A SARS-CoV-2 infectivity assay was conducted in Vero-E6 and Calu-3 epithelial cell lines. The antiviral effect of hemin, and chloroquine as a control, against SARS-CoV-2 virus infection was quantified by RT-qPCR using specific oligonucleotides for the N gene. Chloroquine induced a marked reduction of viral genome copies in kidney epithelial Vero-E6 cells but not in lung cancer Calu-3 cells. Hemin administration to the culture medium induced a high induction in the expression of the HO-1 gene that was stronger in Vero-E6 macaque-derived cells than in the human Calu-3 cell line. However, hemin treatment did not modify SARS-CoV-2 replication, as measured by viral genome quantification 48 h post-infection for Vero-E6 and 72 h post-infection for the Calu-3 lineages. In conclusion, although exposure to hemin induced strong HO-1 up-regulation, this effect was unable to inhibit or delay the progression of SARS-CoV-2 infection in two epithelial cell lines susceptible to infection.


Asunto(s)
Antivirales/farmacología , Hemo-Oxigenasa 1/metabolismo , Hemina/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19 , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Cloroquina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Pulmón/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
9.
Free Radic Biol Med ; 161: 263-271, 2020 12.
Artículo en Inglés | MEDLINE | ID: covidwho-872071

RESUMEN

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to infect hundred thousands of people every day worldwide. Since it is a novel virus, research continues to update the possible therapeutic targets when new evidence regarding COVID-19 are gathered. This article presents an evidence-based hypothesis that activating the heme oxygenase-1 (HO-1) pathway is a potential target for COVID-19. Interferons (IFNs) have broad-spectrum antiviral activity including against SARS-CoV-2. Induction of HO-1 and increase in the heme catabolism end-product confer antiviral activity. IFN activation results in inhibition of viral replication in various viral infections. COVID-19 induced inflammation as well as acute respiratory distress syndrome (ARDS), and coagulopathies are now known major causes of mortality. A protective role of HO-1 induction in inflammation, inflammation-induced coagulation, and ARDS has been reported. Based on an association of HO-1 promoter polymorphisms and disease severity, we propose an evaluation of the status of these polymorphisms in COVID-19 patients who become severely ill. If an association is established, it might be helpful in identifying patients at high risk. Hence, we hypothesize that HO-1 pathway activation could be a therapeutic strategy against COVID-19 and associated complications.


Asunto(s)
COVID-19/inmunología , Fibrinolíticos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Interferón Tipo I/inmunología , SARS-CoV-2/crecimiento & desarrollo , Antivirales/metabolismo , Coagulación Intravascular Diseminada/prevención & control , Hemo/metabolismo , Hemo-Oxigenasa 1/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , SARS-CoV-2/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
10.
Mol Med ; 26(1): 90, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: covidwho-801066

RESUMEN

Hydrogen sulfide (H2S) is a natural defence against the infections from enveloped RNA viruses and is likely involved also in Covid 19. It was already shown to inhibit growth and pathogenic mechanisms of a variety of enveloped RNA viruses and it was now found that circulating H2S is higher in Covid 19 survivors compared to fatal cases. H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Subjects with a long promoter for the HMOX1 gene, coding for HO-1, are predicted for lower efficiency of this mechanism. SARS-cov-2 exerts ability to attack the heme of hemoglobin and other heme-proteins thus hampering both release and signalling of H2S. Lack of H2S-induced persulfidation of the KATP channels of leucocytes causes adhesion and release of the inflammatory cytokines, lung infiltration and systemic endothelial damage with hyper-coagulability. These events largely explain the sex and age distribution, clinical manifestations and co-morbidities of Covid-19. The understanding of this mechanism may be of guidance in re-evaluating the ongoing therapeutic strategies, with special attention to the interaction with mechanical ventilation, paracetamol and chloroquine use, and in the individuation of genetic traits causing increased susceptibility to the disruption of these physiologic processes and to a critical Covid 19. Finally, an array of therapeutic interventions with the potential to clinically modulate the HO-1/CO/H2S axis is already available or under development. These include CO donors and H2S donors and a boost to the endogenous production of H2S is also possible.


Asunto(s)
Infecciones por Coronavirus/inmunología , Sulfuro de Hidrógeno/metabolismo , Neumonía Viral/inmunología , COVID-19 , Monóxido de Carbono/metabolismo , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/terapia , Predisposición Genética a la Enfermedad , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/metabolismo , Neumonía Viral/terapia , Factores de Riesgo
11.
Int J Mol Sci ; 21(17)2020 Sep 03.
Artículo en Inglés | MEDLINE | ID: covidwho-742801

RESUMEN

The coronavirus disease of 2019 (COVID-19) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global pandemic with increasing incidence and mortality rates. Recent evidence based on the cytokine profiles of severe COVID-19 cases suggests an overstimulation of macrophages and monocytes associated with reduced T-cell abundance (lymphopenia) in patients infected with SARS-CoV-2. The SARS-CoV-2 open reading frame 3 a (ORF3a) protein was found to bind to the human HMOX1 protein at a high confidence through high-throughput screening experiments. The HMOX1 pathway can inhibit platelet aggregation, and can have anti-thrombotic and anti-inflammatory properties, amongst others, all of which are critical medical conditions observed in COVID-19 patients. Here, we review the potential of modulating the HMOX1-ORF3a nexus to regulate the innate immune response for therapeutic benefits in COVID-19 patients. We also review other potential treatment strategies and suggest novel synthetic and natural compounds that may have the potential for future development in clinic.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Hemo-Oxigenasa 1/metabolismo , Neumonía Viral/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Animales , Antivirales/uso terapéutico , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Hemo-Oxigenasa 1/genética , Humanos , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Unión Proteica , Proteínas Viroporinas
12.
Med Hypotheses ; 144: 110242, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-739959

RESUMEN

The outbreak of coronavirus disease 2019 (COVID-19) requires urgent need for effective treatment. Severe COVID-19 is characterized by a cytokine storm syndrome with subsequent multiple organ failure (MOF) and acute respiratory distress syndrome (ARDS), which may lead to intensive care unit and increased risk of death. While awaiting a vaccine, targeting COVID-19-induced cytokine storm syndrome appears currently as the efficient strategy to reduce the mortality of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The stress-responsive enzyme, heme oxygenase-1 (HO-1) is largely known to protect against inflammatory response in animal models. HO-1 is induced by hemin, a well-tolerated molecule, used for decades in the treatment of acute intermittent porphyria. Experimental studies showed that hemin-induced HO-1 mitigates cytokine storm and lung injury in mouse models of sepsis and renal ischemia-reperfusion injury. Furthermore, HO-1 may also control numerous viral infections by inhibiting virus replication. In this context, we suggest the hypothesis that HO-1 cytoprotective pathway might be a promising target to control SARS-CoV-2 infection and mitigate COVID-19-induced cytokine storm and subsequent ARDS.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/metabolismo , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Hemo-Oxigenasa 1/metabolismo , Síndrome de Dificultad Respiratoria/fisiopatología , Animales , Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Vacunas contra la COVID-19 , Cuidados Críticos , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/metabolismo , Hemina/metabolismo , Humanos , Inflamación , Interleucina-6/metabolismo , Modelos Teóricos , Polimorfismo Genético , Síndrome de Dificultad Respiratoria/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA