Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Eur J Clin Invest ; 51(11): e13661, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34324704

RESUMEN

BACKGROUND: Unravelling autoimmune targets triggered by SARS-CoV-2 infection may provide crucial insights into the physiopathology of the disease and foster the development of potential therapeutic candidate targets and prognostic tools. We aimed at determining (a) the association between anti-SARS-CoV-2 and anti-apoA-1 humoral response and (b) the degree of linear homology between SARS-CoV-2, apoA-1 and Toll-like receptor 2 (TLR2) epitopes. DESIGN: Bioinformatics modelling coupled with mimic peptides engineering and competition experiments were used to assess epitopes sequence homologies. Anti-SARS-CoV-2 and anti-apoA-1 IgG as well as cytokines were assessed by immunoassays on a case-control (n = 101), an intensive care unit (ICU; n = 126) and a general population cohort (n = 663) with available samples in the pre and post-pandemic period. RESULTS: Using bioinformatics modelling, linear sequence homologies between apoA-1, TLR2 and Spike epitopes were identified but without experimental evidence of cross-reactivity. Overall, anti-apoA-1 IgG levels were higher in COVID-19 patients or anti-SARS-CoV-2 seropositive individuals than in healthy donors or anti-SARS-CoV-2 seronegative individuals (P < .0001). Significant and similar associations were noted between anti-apoA-1, anti-SARS-CoV-2 IgG, cytokines and lipid profile. In ICU patients, anti-SARS-CoV-2 and anti-apoA-1 seroconversion rates displayed similar 7-day kinetics, reaching 82% for anti-apoA-1 seropositivity. In the general population, SARS-CoV-2-exposed individuals displayed higher anti-apoA-1 IgG seropositivity rates than nonexposed ones (34% vs 16.8%; P = .004). CONCLUSION: COVID-19 induces a marked humoral response against the major protein of high-density lipoproteins. As a correlate of poorer prognosis in other clinical settings, such autoimmunity signatures may relate to long-term COVID-19 prognosis assessment and warrant further scrutiny in the current COVID-19 pandemic.


Asunto(s)
Anticuerpos Antivirales/inmunología , Apolipoproteína A-I/inmunología , Autoanticuerpos/inmunología , COVID-19/inmunología , Citocinas/inmunología , Inmunidad Humoral/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Apolipoproteína A-I/química , Biología Computacional , Epítopos/química , Femenino , Humanos , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Péptidos , SARS-CoV-2 , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/química , Receptor Toll-Like 2/química , Receptor Toll-Like 2/inmunología , Adulto Joven
2.
Future Microbiol ; 16: 607-613, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33998267

RESUMEN

Apolipoprotein A-I (apo A-I) represents the main component of the Trypanosome lytic factor (TLF) which contributes to the host innate immunity against Trypanosoma and Leishmania. These parasites use complex and multiple strategies such as molecular mimicry to evade or subvert the host immune system. Previous studies have highlighted the adaptation mechanisms of TLF-resistant Trypanosoma species. These data might support the hypothesis that Leishmania parasites (amastigote forms in macrophages) might express apo A-I to bypass and escape from TLF action as a component of the host innate immune responses. The anti-inflammatory property of apo A-I is another mechanism that supports our idea that apo A-I may play a role in Leishmania parasites allowing them to bypass the host innate immune system.


Asunto(s)
Apolipoproteína A-I/inmunología , Leishmania/inmunología , Leishmaniasis/inmunología , Proteínas Protozoarias/inmunología , Humanos , Evasión Inmune , Inmunidad Innata , Lipoproteínas HDL/inmunología , Macrófagos/inmunología , Macrófagos/parasitología , Imitación Molecular
3.
J Virol ; 95(13): e0197420, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33827950

RESUMEN

Dengue is a mosquito-borne infectious disease that is highly endemic in tropical and subtropical countries. Symptomatic patients can rapidly progress to severe conditions of hemorrhage, plasma extravasation, and hypovolemic shock, which leads to death. The blood tests of patients with severe dengue typically reveal low levels of high-density lipoprotein (HDL), which is responsible for reverse cholesterol transport (RCT) and regulation of the lipid composition in peripheral tissues. It is well known that dengue virus (DENV) depends on membrane cholesterol rafts to infect and to replicate in mammalian cells. Here, we describe the interaction of DENV nonstructural protein 1 (NS1) with apolipoprotein A1 (ApoA1), which is the major protein component of HDL. NS1 is secreted by infected cells and can be found circulating in the serum of patients with the onset of symptoms. NS1 concentrations in plasma are related to dengue severity, which is attributed to immune evasion and an acute inflammatory response. Our data show that the DENV NS1 protein induces an increase of lipid rafts in noninfected cell membranes and enhances further DENV infection. We also show that ApoA1-mediated lipid raft depletion inhibits DENV attachment to the cell surface. In addition, ApoA1 is able to neutralize NS1-induced cell activation and to prevent NS1-mediated enhancement of DENV infection. Furthermore, we demonstrate that the ApoA1 mimetic peptide 4F is also capable of mediating lipid raft depletion to control DENV infection. Taken together, our results suggest the potential of RCT-based therapies for dengue treatment. These results should motivate studies to assess the importance of RCT in DENV infection in vivo. IMPORTANCE DENV is one of the most relevant mosquito-transmitted viruses worldwide, infecting more than 390 million people every year and leading to more than 20 thousand deaths. Although a DENV vaccine has already been approved, its potential side effects have hampered its use in large-scale immunizations. Therefore, new treatment options are urgently needed to prevent disease worsening or to improve current clinical management of severe cases. In this study, we describe a new interaction of the NS1 protein, one of the major viral components, with a key component of HDL, ApoA1. This interaction seems to alter membrane susceptibility to virus infection and modulates the mechanisms triggered by DENV to evade the immune response. We also propose the use of a mimetic peptide named 4F, which was originally developed for atherosclerosis, as a potential therapy for relieving DENV symptoms.


Asunto(s)
Apolipoproteína A-I/inmunología , Virus del Dengue/metabolismo , Evasión Inmune/inmunología , Microdominios de Membrana/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Antivirales/farmacología , Línea Celular , Colesterol/metabolismo , Dengue/patología , Humanos , Inflamación/prevención & control , Ratones , Péptidos/farmacología , Células RAW 264.7 , Acoplamiento Viral/efectos de los fármacos
4.
J Intern Med ; 285(1): 49-58, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30028049

RESUMEN

OBJECTIVE: IgG antibodies against apolipoprotein A-I (ApoA-I) have been found to be elevated in subjects from the general population with clinically manifest cardiovascular disease and in myocardial infarction patients with an adverse prognosis. Here, we investigated whether these antibodies are prospectively associated with carotid artery disease progression and with the risk for first-time cardiovascular events in individuals with no previous history of cardiovascular disease. APPROACH AND RESULTS: We selected 383 subjects from the cardiovascular cohort of Malmö Diet and Cancer study who suffered a coronary event during a median follow-up period of 15.4 (10.3-16.4) years and 395 age- and sex-matched controls. None of the study participants had a previous history of coronary artery disease or stroke. Anti-ApoA-I IgG were measured by ELISA in serum samples collected at baseline. Intima-media thickness (IMT) was measured in the common carotid artery and in the carotid bifurcation at baseline and after 15.9 (±1.5) years. We found no associations between anti-ApoA-I IgG and carotid artery IMT at baseline or with IMT progression during follow-up. In Cox proportional hazards analyses adjusted for traditional cardiovascular risk factors, the hazard ratio (HR 95%CI) for the primary outcome, incident coronary events, was 0.97 (0.75-1.25), P = 0.782, in subjects with anti-ApoA-I IgG within the highest tertile compared with the lowest tertile. Similarly, we did not find any associations with the secondary outcome, incident first-time stroke. CONCLUSIONS: Serum autoantibodies against ApoA-I do not correlate with disease progression and adverse events in cardiovascular disease-free individuals from the general population.


Asunto(s)
Apolipoproteína A-I/inmunología , Enfermedades Cardiovasculares/inmunología , Enfermedades de las Arterias Carótidas/inmunología , Inmunoglobulina G/inmunología , Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Grosor Intima-Media Carotídeo , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Suecia , Ultrasonografía
5.
Hepatol Int ; 12(1): 17-25, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29423541

RESUMEN

BACKGROUND/PURPOSE: One to three per cent of the world's population has hepatitis C virus (HCV) infection, which is not only a major cause of liver disease and cancer but also associated with an increased risk of atherosclerosis, despite an ostensibly favourable lipid profile. Autoantibodies are frequent in HCV infection and emerging evidence shows that autoantibodies could be valuable for cardiovascular disease (CVD) risk stratification. This study investigated a novel independent biomarker of CVD, autoantibodies to apolipoprotein A-1 (anti-apoA-1 IgG) and lipids in patients with chronic HCV before, during and after direct-acting anti-viral (DAA) therapy. METHODS: Eighty-nine blinded serum samples from 27 patients with advanced chronic HCV were assayed for lipids and anti-apoA-1 IgG by ELISA. RESULTS: Pre-treatment HCV viral load correlated with high-density lipoprotein cholesterol (HDL-C, r = 0.417; p = 0.042) and negatively with apolipoprotein (apo)B (r = - 0.497; p = 0.013) and markers of CVD risk, the apoB/apoA-1 ratio (r = - 0.490; p = 0.015) and triglyceride level (TG)/HDL-C ratio (r = - 0.450; p = 0.031). Fourteen (52%) of 27 patients had detectable anti-apoA-1 IgG autoantibodies pre-treatment; only two became undetectable with virological cure. Autoantibody-positive sera had lower apoA-1 (p = 0.012), HDL-C (p = 0.009) and total cholesterol (p = 0.006) levels. CONCLUSIONS: This is the first report of the presence of an emerging biomarker for atherosclerosis, anti-apoA-1 IgG, in some patients with HCV infection. It may be induced by apoA-1 on the surface of HCV lipoviral particles. The autoantibodies inversely correlate with apoA-1 and HDL levels and may render HDL dysfunctional. Whether these hypothesis-generating findings have clinical implications in HCV patients requires further study.


Asunto(s)
Apolipoproteína A-I/inmunología , Autoanticuerpos/sangre , Biomarcadores/sangre , Enfermedad de la Arteria Coronaria/sangre , Hepatitis C/inmunología , Anciano , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/inmunología , Femenino , Hepacivirus/inmunología , Hepatitis C/virología , Humanos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Carga Viral
6.
J Infect ; 76(2): 186-195, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29198606

RESUMEN

OBJECTIVES: To determine the existence of autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) in HIV patients and explore their association with biological features of HIV infection and different inflammatory biomarkers. We also evaluated their impact on CD4+ lymphocytes survival. METHODS: Anti-apoA-1 IgG plasma levels were assessed by ELISA in 237 HIV positive patients from a national prospective cohort with no current lipid-lowering therapy. RESULTS: 58% of patients were found positive for anti-apoA-1 IgG and were associated with lower CD4+ counts, but higher viremia and systemic inflammation. Logistic regression analyses indicated that high anti-apoA-1 IgG levels were associated with a 16-fold increased risk of displaying low CD4+ levels, independent of HIV RNA levels and treatment (adjusted Odds ratio [OR]:16.1, 95% Confidence Interval [95%CI]:1.80-143.6; p = 0.01), and a 6-fold increased risk of having a detectable viremia, independent of antiretroviral treatment (OR:5.47; 95% CI:1.63-18.36; p = 0.006). In vitro, anti-apoA-1 IgG induced dose and time-dependent CD4+ apoptosis that was increased by exposure to HIV RNA. CONCLUSIONS: In HIV patients, anti-apoA-1 IgG levels are associated with low CD4+ counts, high viremia and a pro-inflammatory systemic profile. Anti-apoA-1 IgG can promote CD4+ lymphocyte apoptosis via undefined pathways.


Asunto(s)
Apolipoproteína A-I/inmunología , Autoanticuerpos/inmunología , Infecciones por VIH/inmunología , Inmunoglobulina G/inmunología , Síndromes de Inmunodeficiencia/etiología , Inflamación/sangre , Adulto , Apoptosis , Autoanticuerpos/sangre , Biomarcadores/sangre , Linfocitos T CD4-Positivos/virología , Línea Celular , Femenino , VIH , Humanos , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Estudios Prospectivos , ARN Viral/inmunología , Factores de Riesgo
7.
Biosci Rep ; 37(1)2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28093456

RESUMEN

Activating transcription factor 3 (ATF3) is a TLR-induced repressor that plays an important role in the inhibition of specific inflammatory signals. We previously constructed recombinant high density lipoproteins (rHDL) (including rHDLWT, rHDLM, rHDL228 and rHDL74) and found that rHDL74 had a strong anti-inflammatory ability. In the present study, we investigate the roles of recombinant apolipoprotein A-I (ApoA-I) (rHDLWT) and its cysteine mutant HDLs (rHDLM, rHDL228 and rHDL74) on ATF3 function in RAW264.7 cells stimulated by lipopolysaccharide. Our results showed that compared with the LPS group, rHDL74 can decrease the level of TNF-α and IL-6, whereas rHDL228 increases their expression levels. RT-PCR and Western blotting results showed that compared with the LPS group, rHDL74, rHDLWT and rHDLM can markedly increase the expression level of ATF3, whereas the level of ATF3 decreases in the rHDL228 group. In summary, the different anti-inflammatory mechanisms of the ApoA-I cysteine mutants might be associated with the regulation of ATF3 level.


Asunto(s)
Factor de Transcripción Activador 3/inmunología , Apolipoproteína A-I/inmunología , Macrófagos/inmunología , Animales , Apolipoproteína A-I/genética , Cisteína/genética , Cisteína/inmunología , Humanos , Interleucina-6/inmunología , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Ratones , Mutación Puntual , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Factor de Necrosis Tumoral alfa/inmunología
8.
Eur J Clin Invest ; 46(9): 805-17, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27490973

RESUMEN

BACKGROUND: Autoantibodies to apolipoprotein A-1 (anti-ApoA-1 IgG) were shown to predict major adverse cardiovascular events and promote atherogenesis. However, their potential relationship with clinical disability and ischaemic lesion volume after acute ischaemic stroke (AIS) remains unexplored. MATERIALS AND METHODS: We included n = 76 patients admitted for AIS and we investigated whether baseline serum anti-ApoA-1 IgG levels could predict (i) AIS-induced clinical disability [assessed by the modified Rankin Scale (mRS)], and (ii) AIS-related ischaemic lesion volume [assessed by Computed Tomography (CT)]. We also evaluated the possible pro-apoptotic and pro-necrotic effects of anti-ApoA-1 IgG on human astrocytoma cell line (U251) using flow cytometry. RESULTS: High levels of anti-ApoA-1 IgG were retrieved in 15·8% (12/76) of patients. Increased baseline levels of anti-ApoA-1 IgG were independently correlated with worse mRS [ß = 0·364; P = 0·002; adjusted odds ratio (OR): 1·05 (95% CI 1·01-1·09); P = 0·017] and CT-assessed ischaemic lesion volume [ß = 0·333; P < 0·001; adjusted OR: 1·06 (95% CI 1·01-1·12); P = 0·048] at 3 months. No difference in baseline clinical, biochemical and radiological characteristics was observed between patients with high vs. low levels of anti-ApoA-1 IgG. Incubating human astrocytoma cells with anti-ApoA-1 IgG dose dependently induced necrosis and apoptosis of U251 cells in vitro. CONCLUSION: Anti-ApoA-1 IgG serum levels at AIS onset are associated with poorer clinical recovery and worse brain lesion volume 3 months after AIS. These observations could be partly explained by the deleterious effect of anti-ApoA-1 IgG on human brain cell survival in vitro and may have clinical implication in the prediction of poor outcome in AIS.


Asunto(s)
Apolipoproteína A-I/inmunología , Autoanticuerpos/inmunología , Inmunoglobulina G/inmunología , Accidente Cerebrovascular/inmunología , Anciano , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Autoanticuerpos/farmacología , Línea Celular Tumoral , Femenino , Citometría de Flujo , Estudios de Seguimiento , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Técnicas In Vitro , Receptores de Lipopolisacáridos/efectos de los fármacos , Receptores de Lipopolisacáridos/metabolismo , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Necrosis , Oportunidad Relativa , Proyectos Piloto , Pronóstico , Estudios Prospectivos , Recuperación de la Función , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/fisiopatología , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Tomografía Computarizada por Rayos X
9.
Thromb Haemost ; 116(3): 554-64, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27356567

RESUMEN

Humoral autoimmune-mediated inflammation plays a role in atherogenesis, and potentially in arterial thrombosis. Anti-apolipoprotein A-1 (apoA-1) IgG have been reported to represent emergent mediators of atherogenesis through Toll-like receptors (TLR) 2, 4 and CD14 signalling. We investigated the role of anti-apoA-1 IgG on tissue factor (TF) expression and activation, a key coagulation regulator underlying atherothrombosis. Atherothrombosis features were determined by immunohistochemical TF staining of human carotid biopsies derived from patients with severe carotid stenosis undergoing elective surgery (n=176), and on aortic roots of different genetic backgrounds mice (ApoE-/-; TLR2-/-ApoE-/- and TLR4-/-ApoE-/-) exposed to passive immunisation with anti-apoA-1 IgG. Human serum levels of anti-apoA-1 IgG were measured by ELISA. In vitro, on human-monocyte-derived-macrophages (HMDM) the anti-apoA-1 IgG increased TF expression and activity were analysed by FACS and chromogenic assays in presence of different pharmacological inhibitors. Human serum anti-apoA-1 IgG levels significantly correlated to intraplaque TF expression in carotid biopsies (r=0.31, p<0.001), which was predictive of clinically symptomatic lesions. On HMDM, anti-apoA-1 IgG induced a TLR2, 4 and CD14-dependent increase in TF expression and activity, involving NF-kappaB and a c-Jun N-terminal kinase-dependent AP-1 transcription factors. In ApoE-/- mice, anti-apoA-1 IgG passive immunisation significantly enhanced intraplaque TF expression when compared to control IgG. This effect was lost in both TLR2-/-ApoE-/- and TLR4-/-ApoE-/- mice. These results demonstrate that anti-apoA-1 IgG are associated with TF expression in human atherosclerotic plaques, induce TF expression in vitro and in vivo through TLR2 and 4 signalling, supporting a possible causal relationship between anti-apoA-1 IgG and atherothrombosis.


Asunto(s)
Apolipoproteína A-I/antagonistas & inhibidores , Apolipoproteína A-I/inmunología , Autoanticuerpos/sangre , Trombosis/etiología , Trombosis/inmunología , Anciano , Anciano de 80 o más Años , Animales , Estenosis Carotídea/sangre , Estenosis Carotídea/etiología , Estenosis Carotídea/inmunología , Estudios de Cohortes , Femenino , Humanos , Inmunoglobulina G/sangre , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Ratones Noqueados para ApoE , Placa Aterosclerótica/sangre , Placa Aterosclerótica/etiología , Placa Aterosclerótica/inmunología , Estudios Prospectivos , Tromboplastina/metabolismo , Trombosis/sangre , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
10.
Thromb Haemost ; 116(4): 764-71, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27384400

RESUMEN

We aimed to determine the association between autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) and prevalent cardiovascular (CV) disease (CVD) as well as markers of CV risk in the general population. Cross-sectional data were obtained from 6649 subjects (age 52.6 ± 10.7 years, 47.4 % male) of the population-based CoLaus study. CVD was defined as myocardial infarction, angina pectoris, percutaneous revascularisation or bypass grafting for ischaemic heart disease stroke or transient ischaemic attack, and was assessed according to standardised medical records. Anti-apoA-1 IgG and biological markers were measured by ELISA and conventional automated techniques, respectively. Prevalence of high anti-apoA-1 IgG levels in the general population was 19.9 %. Presence of anti-apoA-1 IgG was significantly associated with CVD [odds ratio 1.34, 95 % confidence interval (1.05-1.70), p=0.018], independently of established CV risk factors (CVRFs) including age, sex, hypertension, smoking, diabetes, low and high-density lipoprotein cholesterol levels. The n=455 (6.8 %) study participants with a history of CVD (secondary prevention subgroup) presented higher median anti-ApoA-1 IgG values compared with subjects without CVD (p=0.029). Among patients in the secondary prevention subgroup, those with positive anti-apoA-1 IgG levels had lower HDL (p=0.002) and magnesium (p=0.001) levels, but increased uric acid and high-sensitivity C-reactive protein levels (p=0.022, and p<0.001, respectively) compared to patients with negative anti-apoA-1 IgG levels. In conclusion, anti-apoA-1 IgG levels are independently associated with CVD in the general population and also related to CV biomarkers in secondary prevention. These findings indicate that anti-apoA-1 IgG may represent a novel CVRF and need further study in prospective cohorts.


Asunto(s)
Apolipoproteína A-I/inmunología , Autoanticuerpos/sangre , Enfermedades Cardiovasculares/sangre , Adulto , Enfermedades Cardiovasculares/inmunología , Estudios Transversales , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad
11.
J Immunol ; 197(2): 611-9, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27316682

RESUMEN

Synthetic amphipathic helical peptides (SAHPs) designed as apolipoprotein A-I mimetics are known to bind to class B scavenger receptors (SR-Bs), SR-BI, SR-BII, and CD36, receptors that mediate lipid transport and facilitate pathogen recognition. In this study, we evaluated SAHPs, selected for targeting human CD36, by their ability to attenuate LPS-induced inflammation, endothelial barrier dysfunction, and acute lung injury (ALI). L37pA, which targets CD36 and SR-BI equally, inhibited LPS-induced IL-8 secretion and barrier dysfunction in cultured endothelial cells while reducing lung neutrophil infiltration by 40% in a mouse model of LPS-induced ALI. A panel of 20 SAHPs was tested in HEK293 cell lines stably transfected with various SR-Bs to identify SAHPs with preferential selectivity toward CD36. Among several SAHPs targeting both SR-BI/BII and CD36 receptors, ELK-B acted predominantly through CD36. Compared with L37pA, 5A, and ELK SAHPs, ELK-B was most effective in reducing the pulmonary barrier dysfunction, neutrophil migration into the lung, and lung inflammation induced by LPS. We conclude that SAHPs with relative selectivity toward CD36 are more potent at inhibiting acute pulmonary inflammation and dysfunction. These data indicate that therapeutic strategies using SAHPs targeting CD36, but not necessarily mimicking all apolipoprotein A-I functions, may be considered a possible new treatment approach for inflammation-induced ALI and pulmonary edema.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Antiinflamatorios/farmacología , Antígenos CD36/antagonistas & inhibidores , Inflamación/inmunología , Lesión Pulmonar Aguda/patología , Animales , Apolipoproteína A-I/inmunología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamación/patología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología
12.
Chest ; 150(2): 283-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27327118

RESUMEN

New treatments are needed for patients with asthma who are refractory to standard therapies, such as individuals with a phenotype of "type 2-low" inflammation. This important clinical problem could potentially be addressed by the development of apolipoprotein A-I (apoA-I) mimetic peptides. ApoA-I interacts with its cellular receptor, the ATP-binding cassette subfamily A, member 1 (ABCA1), to facilitate cholesterol efflux out of cells to form nascent high-density lipoprotein particles. The ability of the apoA-I/ABCA1 pathway to promote cholesterol efflux from cells that mediate adaptive immunity, such as antigen-presenting cells, can attenuate their function. Data from experimental murine models have shown that the apoA-I/ABCA1 pathway can reduce neutrophilic airway inflammation, primarily by suppressing the production of granulocyte-colony stimulating factor. Furthermore, administration of apoA-I mimetic peptides to experimental murine models of allergic asthma has decreased both neutrophilic and eosinophilic airway inflammation, as well as airway hyperresponsiveness and mucous cell metaplasia. Higher serum levels of apoA-I have also been associated with less severe airflow obstruction in patients with asthma. Collectively, these results suggest that the apoA-I/ABCA1 pathway may have a protective effect in asthma, and support the concept of advancing inhaled apoA-I mimetic peptides to clinical trials that can assess their safety and effectiveness. Thus, we propose that the development of inhaled apoA-I mimetic peptides as a new treatment could represent a clinical advance for patients with severe asthma who are unresponsive to other therapies.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/inmunología , Apolipoproteína A-I/inmunología , Asma/inmunología , Hiperreactividad Bronquial/inmunología , Inflamación/inmunología , Transportador 1 de Casete de Unión a ATP/metabolismo , Administración por Inhalación , Apolipoproteína A-I/metabolismo , Asma/tratamiento farmacológico , Asma/metabolismo , Transporte Biológico , Hiperreactividad Bronquial/tratamiento farmacológico , Hiperreactividad Bronquial/metabolismo , Colesterol/metabolismo , Descubrimiento de Drogas , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipoproteínas HDL/metabolismo , Redes y Vías Metabólicas , Terapia Molecular Dirigida , Péptidos
13.
J Immunol ; 195(10): 4685-98, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466956

RESUMEN

Apolipoprotein (Apo)A-I, the major lipid-binding protein of high-density lipoprotein, can prevent autoimmunity and suppress inflammation in hypercholesterolemic mice by attenuating lymphocyte cholesterol accumulation and removing tissue-oxidized lipids. However, whether ApoA-I mediates immune-suppressive or anti-inflammatory effects under normocholesterolemic conditions and the mechanisms involved remain unresolved. We transferred bone marrow from systemic lupus erythematosus (SLE)-prone Sle123 mice into normal, ApoA-I-knockout (ApoA-I(-/-)) and ApoA-I-transgenic (ApoA-I(tg)) mice. Increased ApoA-I in ApoA-I(tg) mice suppressed CD4(+) T and B cell activation without changing lymphocyte cholesterol levels or reducing major ApoA-I-binding oxidized fatty acids. Unexpectedly, oxidized fatty acid peroxisome proliferator-activated receptor γ ligands 13- and 9-hydroxyoctadecadienoic acid were increased in lymphocytes of autoimmune ApoA-I(tg) mice. ApoA-I reduced Th1 cells independently of changes in CD4(+)Foxp3(+) regulatory T cells or CD11c(+) dendritic cell activation and migration. Follicular helper T cells, germinal center B cells, and autoantibodies were also lower in ApoA-I(tg) mice. Transgenic ApoA-I also improved SLE-mediated glomerulonephritis. However, ApoA-I deficiency did not have the opposite effects on autoimmunity or glomerulonephritis, possibly as the result of compensatory increases in ApoE on high-density lipoprotein. We conclude that, although compensatory mechanisms prevent the proinflammatory effects of ApoA-I deficiency in normocholesterolemic mice, increasing ApoA-I can attenuate lymphocyte activation and autoimmunity in SLE independently of cholesterol transport, possibly through oxidized fatty acid peroxisome proliferator-activated receptor γ ligands, and it can reduce renal inflammation in glomerulonephritis.


Asunto(s)
Apolipoproteína A-I/metabolismo , Autoinmunidad/inmunología , Colesterol/metabolismo , Nefritis Lúpica/inmunología , Activación de Linfocitos/inmunología , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/inmunología , Apolipoproteínas E/metabolismo , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoinmunidad/genética , Linfocitos B/inmunología , Trasplante de Médula Ósea , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Cromatografía de Gases y Espectrometría de Masas , Ácidos Linoleicos/metabolismo , Lipoproteínas HDL/inmunología , Nefritis Lúpica/genética , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/metabolismo , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
14.
Endocrinology ; 156(12): 4707-19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26393305

RESUMEN

In vitro and animal studies point to autoantibodies against apolipoprotein A-1 (anti-apoA-1 IgG) as possible mediators of cardiovascular (CV) disease involving several mechanisms such as basal heart rate interference mediated by a mineralocorticoid receptor-dependent L-type calcium channel activation, and a direct pro-inflammatory effect through the engagement of the toll-like receptor (TLR) 2/CD14 complex. Nevertheless, the possible implication of these receptors in the pro-arrhythmogenic effect of anti-apoA-1 antibodies remains elusive. We aimed at determining whether CD14 and TLRs could mediate the anti-apoA-1 IgG chronotropic response in neonatal rat ventricular cardiomyocytes (NRVC). Blocking CD14 suppressed anti-apoA-1 IgG binding to NRVC and the related positive chronotropic response. Anti-apoA-1 IgG alone induced the formation of a TLR2/TLR4/CD14 complex, followed by the phosphorylation of Src, whereas aldosterone alone promoted the phosphorylation of Akt by phosphatidylinositol 3-kinase (PI3K), without affecting the chronotropic response. In the presence of both aldosterone and anti-apoA-1 IgG, the localization of TLR2/TLR4/CD14 was increased in membrane lipid rafts, followed by PI3K and Src activation, leading to an L-type calcium channel-dependent positive chronotropic response. Pharmacological inhibition of the Src pathway led to the decrease of L-type calcium channel activity and abrogated the NRVC chronotropic response. Activation of CD14 seems to be a key regulator of the mineralocorticoid receptor-dependent anti-apoA-1 IgG positive chronotropic effect on NRVCs, involving relocation of the CD14/TLR2/TLR4 complex into lipid rafts followed by PI3K and Src-dependent L-type calcium channel activation.


Asunto(s)
Apolipoproteína A-I/inmunología , Autoanticuerpos/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Inmunoglobulina G/inmunología , Receptores de Lipopolisacáridos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Receptores de Mineralocorticoides/efectos de los fármacos , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/efectos de los fármacos , Ventrículos Cardíacos/citología , Receptores de Lipopolisacáridos/inmunología , Miocitos Cardíacos/inmunología , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/efectos de los fármacos , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Ratas , Ratas Wistar , Receptores de Mineralocorticoides/inmunología , Transducción de Señal , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología
15.
PLoS One ; 10(7): e0132780, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26177543

RESUMEN

BACKGROUND: Cardiovascular disease (CVD) is the leading cause of death worldwide and new approaches for both diagnosis and treatment are required. Autoantibodies directed against apolipoprotein A-I (ApoA-I) represent promising biomarkers for use in risk stratification of CVD and may also play a direct role in pathogenesis. METHODOLOGY: To characterize the anti-ApoA-I autoantibody response, we measured the immunoreactivity to engineered peptides corresponding to the different alpha-helical regions of ApoA-I, using plasma from acute chest pain cohort patients known to be positive for anti-ApoA-I autoantibodies. PRINCIPAL FINDINGS: Our results indicate that the anti-ApoA-I autoantibody response is strongly biased towards the C-terminal alpha-helix of the protein, with an optimized mimetic peptide corresponding to this part of the protein recapitulating the diagnostic accuracy for an acute ischemic coronary etiology (non-ST segment elevation myocardial infarction and unstable angina) obtainable using intact endogenous ApoA-I in immunoassay. Furthermore, the optimized mimetic peptide strongly inhibits the pathology-associated capacity of anti-ApoA-I antibodies to elicit proinflammatory cytokine release from cultured human macrophages. CONCLUSIONS: In addition to providing a rationale for the development of new approaches for the diagnosis and therapy of CVD, our observations may contribute to the elucidation of how anti-ApoA-I autoantibodies are elicited in individuals without autoimmune disease.


Asunto(s)
Apolipoproteína A-I/química , Apolipoproteína A-I/inmunología , Autoanticuerpos/inmunología , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/terapia , Secuencia de Aminoácidos , Enfermedades Cardiovasculares/sangre , Dicroismo Circular , Humanos , Proteínas Inmovilizadas/metabolismo , Inmunoglobulina G/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/farmacología , Datos de Secuencia Molecular , Péptidos/química , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Factor de Necrosis Tumoral alfa/farmacología
16.
PLoS One ; 10(5): e0125112, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25970596

RESUMEN

First-episode psychosis (FEP) is associated with inflammatory and brain structural changes, but few studies have investigated whether systemic inflammation associates with brain structural changes in FEP. Thirty-seven FEP patients (median 27 days on antipsychotic medication), and 19 matched controls were recruited. Serum levels of 38 chemokines and cytokines, and cardiovascular risk markers were measured at baseline and 2 months later. We collected T1- and diffusion-weighted MRIs with a 3 T scanner from the patients at baseline. We analyzed the association of psychosis-related inflammatory markers with gray and white matter (WM) volume using voxel-based morphometry and WM diffusion using tract-based spatial statistics with whole-brain and region-of-interest (ROI) analyses. FEP patients had higher CCL22 and lower TGFα, CXCL1, CCL7, IFN-α2 and ApoA-I than controls. CCL22 decreased significantly between baseline and 2 months in patients but was still higher than in controls. The association between inflammatory markers and FEP remained significant after adjusting for age, sex, smoking and BMI. We did not observe a correlation of inflammatory markers with any symptoms or duration of antipsychotic treatment. Baseline CCL22 levels correlated negatively with WM volume and positively with mean diffusivity and radial diffusivity bilaterally in the frontal lobes in ROI analyses. Decreased serum level of ApoA-I was associated with smaller volume of the medial temporal WM. In whole-brain analyses, CCL22 correlated positively with mean diffusivity and radial diffusivity, and CXCL1 associated negatively with fractional anisotropy and positively with mean diffusivity and radial diffusivity in several brain regions. This is the first report to demonstrate an association between circulating chemokine levels and WM in FEP patients. Interestingly, CCL22 has been previously implicated in autoimmune diseases associated with WM pathology. The results suggest that an altered activation of innate immunity may contribute to WM damage in psychotic disorders.


Asunto(s)
Apolipoproteína A-I/inmunología , Quimiocina CCL22/inmunología , Lóbulo Frontal/inmunología , Inmunidad Innata , Trastornos Psicóticos/inmunología , Sustancia Blanca/inmunología , Adolescente , Adulto , Anisotropía , Antipsicóticos/uso terapéutico , Apolipoproteína A-I/sangre , Estudios de Casos y Controles , Quimiocina CCL22/sangre , Quimiocina CCL7/sangre , Quimiocina CCL7/inmunología , Quimiocina CXCL1/sangre , Quimiocina CXCL1/inmunología , Imagen de Difusión por Resonancia Magnética , Imagen de Difusión Tensora , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Expresión Génica/inmunología , Sustancia Gris/inmunología , Sustancia Gris/metabolismo , Sustancia Gris/patología , Humanos , Procesamiento de Imagen Asistido por Computador , Interferón-alfa/sangre , Interferón-alfa/inmunología , Masculino , Trastornos Psicóticos/sangre , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/patología , Factor de Crecimiento Transformador alfa/sangre , Factor de Crecimiento Transformador alfa/inmunología , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
17.
Biochem Cell Biol ; 93(4): 343-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25994015

RESUMEN

A fusion protein comprising an α-CD20 single chain variable fragment (scFv) antibody, a spacer peptide, and human apolipoprotein (apo) A-I was constructed and expressed in Escherichia coli. The lipid interaction properties intrinsic to apoA-I as well as the antigen recognition properties of the scFv were retained by the chimera. scFv•apoA-I was formulated into nanoscale reconstituted high-density lipoprotein particles (termed nanodisks; ND) and incubated with cultured cells. α-CD20 scFv•apoA-I ND bound to CD20-positive non-Hodgkins lymphoma (NHL) cells (Ramos and Granta) but not to CD20-negative T lymphocytes (i.e., Jurkat). Binding to NHL cells was partially inhibited by pre-incubation with rituximab, a monoclonal antibody directed against CD20. Confocal fluorescence microscopy analysis of Granta cells following incubation with α-CD20 scFv•apoA-I ND formulated with the intrinsically fluorescent hydrophobic polyphenol, curcumin, revealed α-CD20 scFv•apoA-I localizes to the cell surface, while curcumin off-loads and gains entry to the cell. Compared to control incubations, viability of cultured NHL cells was decreased upon incubation with α-CD20 scFv•apoA-I ND harboring curcumin. Thus, formulation of curcumin ND with α-CD20 scFv•apoA-I as the scaffold component confers cell targeting and enhanced bioactive agent delivery, providing a strategy to minimize toxicity associated with chemotherapeutic agents.


Asunto(s)
Antígenos CD20/inmunología , Apolipoproteína A-I/inmunología , Linfoma/terapia , Nanoestructuras , Anticuerpos de Cadena Única/inmunología , Línea Celular Tumoral , Humanos , Linfoma/inmunología , Microscopía Confocal , Proteínas Recombinantes de Fusión/inmunología
18.
Cancer Immunol Immunother ; 64(6): 717-25, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25795134

RESUMEN

Transforming growth factor beta (TGF-ß) promotes tumor growth, invasion and metastasis in established tumors. In this study, we analyzed the effect of overexpressing an anti-TGF-ß peptide fused to apolipoprotein A-I (ApoA-I) as a scaffold molecule. We generated and characterized stable MC38 colon carcinoma clones expressing ApoA-I fused to the anti-TGF-ß peptide P144 and ApoA-I as control cells. We evaluated in vitro the gene expression profile, cell cycle and anchorage-independent growth. The in vivo tumorigenic potential and immunogenicity were analyzed inoculating the MC38 clones into C57BL/6 mice, recombination-activating gene 1 knockout mice or mice deficient in NK cells either subcutaneously or intrasplenically to generate hepatic metastases. While overexpression of ApoA-I had no effect on the parameters analyzed, ApoA-I fused to P144 markedly diminished the tumorigenic capacity and metastatic potential of MC38 in vitro and in vivo, thus generating a highly immunogenic cell line. MC38 cells transfected with ApoA-I fused to P144 triggered memory T cell responses able to eliminate the parental cell line upon re-challenge. In summary, expression of ApoA-I fused to P144 is a novel strategy to modulate TGF-ß in tumor cells. These results highlight the potential of TGF-ß as a target in the development of new antitumor treatments.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Neoplasias del Colon/terapia , Fragmentos de Péptidos/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/inmunología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Femenino , Terapia Genética , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
19.
J Biol Chem ; 289(41): 28249-59, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25170076

RESUMEN

Autoantibodies to apolipoprotein A-I (anti-apoA-I IgG) have been shown to be both markers and mediators of cardiovascular disease, promoting atherogenesis and unstable atherosclerotic plaque. Previous studies have shown that high levels of anti-apoA-I IgGs are independently associated with major adverse cardiovascular events in patients with myocardial infarction. Autoantibody responses to apoA-I can be polyclonal and it is likely that more than one epitope may exist. To identify the specific immunoreactive peptides in apoA-I, we have developed a set of methodologies and procedures to isolate, purify, and identify novel apoA-I endogenous epitopes. First, we generated high purity apoA-I from human plasma, using thiophilic interaction chromatography followed by enzymatic digestion specifically at lysine or arginine residues. Immunoreactivity to the different peptides generated was tested by ELISA using serum obtained from patients with acute myocardial infarction and high titers of autoantibodies to native apoA-I. The immunoreactive peptides were further sequenced by mass spectrometry. Our approach successfully identified two novel immunoreactive peptides, recognized by autoantibodies from patients suffering from myocardial infarction, who contain a high titer of anti-apoA-I IgG. The discovery of these epitopes may open innovative prognostic and therapeutic opportunities potentially suitable to improve current cardiovascular risk stratification.


Asunto(s)
Apolipoproteína A-I/química , Aterosclerosis/inmunología , Autoanticuerpos/sangre , Epítopos/química , Infarto del Miocardio/inmunología , Placa Aterosclerótica/inmunología , Secuencia de Aminoácidos , Apolipoproteína A-I/inmunología , Autoanticuerpos/biosíntesis , Biomarcadores/análisis , Cromatografía de Afinidad , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Expresión Génica , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Datos de Secuencia Molecular , Infarto del Miocardio/sangre , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/patología , Péptidos/química , Péptidos/inmunología , Placa Aterosclerótica/sangre , Placa Aterosclerótica/diagnóstico , Placa Aterosclerótica/patología , Análisis de Secuencia de Proteína
20.
J Biol Chem ; 288(25): 17947-53, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23658016

RESUMEN

HDL has been considered to be a protective factor in sepsis; however, most contributing studies were conducted using the endotoxic animal model, and evidence from clinically relevant septic animal models remains limited and controversial. Furthermore, little is known about the roles of HDL in sepsis other than LPS neutralization. In this study, we employed cecal ligation and puncture (CLP), a clinically relevant septic animal model, and utilized apoA-I knock-out (KO) and transgenic mice to elucidate the roles of HDL in sepsis. ApoA-I-KO mice were more susceptible to CLP-induced septic death as shown by the 47.1% survival of apoA-I-KO mice versus the 76.7% survival of C57BL/6J (B6) mice (p = 0.038). ApoA-I-KO mice had exacerbated inflammatory cytokine production during sepsis compared with B6 mice. Further study indicated that serum from apoA-I-KO mice displayed less capacity for LPS neutralization compared with serum from B6 mice. In addition, apoA-I-KO mice had less LPS clearance, reduced corticosterone generation, and impaired leukocyte recruitment in sepsis. In contrast to apoA-I-KO mice, apoA-I transgenic mice were moderately resistant to CLP-induced septic death compared with B6 mice. In conclusion, our findings reveal multiple protective roles of HDL in CLP-induced sepsis. In addition to its well established role in neutralization of LPS, HDL exerts its protection against sepsis through promoting LPS clearance and modulating corticosterone production and leukocyte recruitment. Our study supports efforts to raise HDL levels as a therapeutic approach for sepsis.


Asunto(s)
Apolipoproteína A-I/inmunología , Infecciones Bacterianas/inmunología , Lipoproteínas HDL/inmunología , Sepsis/inmunología , Animales , Apolipoproteína A-I/genética , Infecciones Bacterianas/complicaciones , Ciego/cirugía , Corticosterona/sangre , Corticosterona/inmunología , Interleucina-6/sangre , Interleucina-6/inmunología , Ligadura , Lipopolisacáridos/sangre , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Lipoproteínas/inmunología , Lipoproteínas/metabolismo , Lipoproteínas HDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infiltración Neutrófila/inmunología , Peritoneo/inmunología , Peritoneo/metabolismo , Punciones , Sepsis/etiología , Sepsis/mortalidad , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA