Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Immunol ; 214(2): 197-208, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37498307

RESUMEN

The interplay between platelets and leukocytes contributes to the pathogenesis of inflammation, thrombosis, and cardiovascular diseases (CVDs) in type 2 diabetes (T2D). Our recent studies described alpha-ketoglutarate (αKG), a Krebs cycle intermediate metabolite as an inhibitor to platelets and leukocytes activation by suppressing phosphorylated-Akt (pAkt) through augmentation of prolyl hydroxylase-2 (PHD2). Dietary supplementation with a pharmacological concentration of αKG significantly inhibited lung inflammation in mice with either SARS-CoV-2 infection or exposed to hypoxia treatment. We therefore investigated if αKG supplementation could suppress hyperactivation of these blood cells and reduce thromboinflammatory complications in T2D. Our study describes that dietary supplementation with αKG (8 mg/100 g body wt. daily) for 7 days significantly reduced the activation of platelets and leukocytes (neutrophils and monocytes), and accumulation of IL1ß, TNFα, and IL6 in peripheral blood of T2D mice. αKG also reduced the infiltration of platelets and leukocytes, and accumulation of inflammatory cytokines in lungs by suppressing pAkt and pP65 signaling. In a cross-sectional investigation, our study also described the elevated platelet-leukocyte aggregates and pro-inflammatory cytokines in circulation of T2D patients. T2D platelets and leukocytes showed an increased aggregation and thrombus formation in vitro. Interestingly, a pre-incubation of T2D blood samples with octyl αKG significantly suppressed the activation of these blood cells and ameliorated aggregate/thrombus formation in vitro. Thus, suggesting a potential therapeutic role of αKG against inflammation, thrombosis, and CVDs in T2D.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Trombosis , Humanos , Ratones , Animales , Ácidos Cetoglutáricos/metabolismo , Estudios Transversales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Activación Plaquetaria , Inflamación/metabolismo , Leucocitos/patología , Plaquetas/patología , Trombosis/tratamiento farmacológico , Trombosis/etiología , Enfermedades Cardiovasculares/patología , Citocinas/metabolismo , Suplementos Dietéticos
2.
IUBMB Life ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38063433

RESUMEN

Diabetes, a chronic metabolic disorder disrupting blood sugar regulation, has emerged as a prominent silent pandemic. Uncontrolled diabetes predisposes an individual to develop fatal complications like cardiovascular disorders, kidney damage, and neuropathies and aggravates the severity of treatable infections. Escalating cases of Type 1 and Type 2 diabetes correlate with a global upswing in diabetes-linked mortality. As a growing global concern with limited preventive interventions, diabetes necessitates extensive research to mitigate its healthcare burden and assist ailing patients. An altered immune system exacerbated by chronic hyperinflammation heightens the susceptibility of diabetic individuals to microbial infections, including notable viruses like SARS-CoV-2, dengue, and influenza. Given such a scenario, we scrutinized the literature and compiled molecular pathways and signaling cascades related to immune compartments in diabetics that escalate the severity associated with the above-mentioned viral infections in them as compared to healthy individuals. The pathogenesis of these viral infections that trigger diabetes compromises both innate and adaptive immune functions and pre-existing diabetes also leads to heightened disease severity. Lastly, this review succinctly outlines available treatments for diabetics, which may hold promise as preventive or supportive measures to effectively combat these viral infections in the former.

3.
Cytokine ; 172: 156380, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37812996

RESUMEN

The endoplasmic reticulum (ER) is crucial for maintaining cellular homeostasis, and synthesis and folding of proteins and lipids. The ER is sensitive to stresses including viral infection that perturb the intracellular energy level and redox state, and accumulating unfolded/misfolded proteins. Viruses including Japanese encephalitis virus (JEV) activates unfolded protein response (UPR) causing ER stress in host immune cells and promotes inflammation and apoptotic cell death. The chemokine receptor CXCR3 has been reported to play important role in the accumulation of inflammatory immune cells and neuronal cell death in several disease conditions. Recently we described the involvement of CXCR3 in regulating inflammation and JEV infection in mice brain. Supplementation with a CXCR3 antagonist AMG487 significantly reduced JEV infection in the mice brain in conjunction with the downregulation of UPR pathway via PERK:eIF2α:CHOP, and decreased mitochondrial ROS generation, inflammation and apoptotic cell death. Alongside, AMG487 treatment improved interferon (IFN)-α/ß synthesis in JEV-infected mice brain. Thus, suggesting a potential therapeutic role of CXCR3 antagonist against JEV infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Animales , Ratones , Encefalitis Japonesa/metabolismo , Estrés del Retículo Endoplásmico , Inflamación/tratamiento farmacológico , Encéfalo/metabolismo
4.
Blood Cells Mol Dis ; 94: 102653, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35180460

RESUMEN

Abnormal coagulation dynamics, including disseminated intravascular coagulopathy, pulmonary embolism, venous thromboembolism and risk of thrombosis are often associated with the severity of COVID-19. However, very little is known about the contribution of platelets in above pathogenesis. In order to decipher the pathophysiology of thrombophilia in COVID-19, we recruited severely ill patients from ICU, based on the above symptoms and higher D-dimer levels, and compared these parameters with their asymptomatic counterparts. Elevated levels of platelet-derived microparticles and platelet-leukocyte aggregates suggested the hyperactivation of platelets in ICU patients. Strikingly, platelet transcriptome analysis showed a greater association of IL-6 and TNF signalling pathways in ICU patients along with higher plasma levels of IL-6 and TNFα. In addition, upregulation of pathways like blood coagulation and hemostasis, as well as inflammation coexisted in platelets of these patients. Further, the increment of necrotic pathway and ROS-metabolic processes in platelets was suggestive of its procoagulant phenotype in ICU patients. This study suggests that higher plasma IL-6 and TNFα may trigger platelet activation and coagulation, and in turn aggravate thrombosis and hypercoagulation in severe COVID-19 patients. Therefore, the elevated IL-6 and TNFα, may serve as potential risk factors for platelet activation and thrombophilia in these patients.


Asunto(s)
COVID-19 , Micropartículas Derivadas de Células , Trombofilia , Plaquetas/metabolismo , COVID-19/complicaciones , Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Humanos , SARS-CoV-2 , Trombofilia/complicaciones , Regulación hacia Arriba
5.
J Clin Immunol ; 39(3): 336-345, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30945073

RESUMEN

A homozygous 83-kb deletion encompassing the genes for complement factor-H-related proteins 1 and 3 (FHR 1, FHR3) is known as a risk factor for some immune inflammatory disorders. However, the functional relevance of this FHR1/3 deletion is relatively unexplored. Globally, healthy populations of all ethnic groups tested show an 8-10% prevalence of homozygosity for this deletion polymorphism. We have begun to compare the peripheral leucocyte phenotype and functionality between FHR1/3-/- and FHR1/3+/+ healthy adult individuals. We report that the two groups show significant differences in their peripheral blood innate leucocyte subset composition, although the adaptive immune subsets are similar between them. Specifically, FHR1/3-/- individuals show higher frequencies of patrolling monocytes and lower frequencies of classical monocytes than FHR1/3+/+ individuals. Similarly, FHR1/3-/- individuals show higher frequencies of plasmacytoid dendritic cells (pDCs) and lower frequencies of myeloid DCs (mDCs) than FHR1/3+/+ individuals. Notably, classical monocytes specifically showed cell-surface-associated factor H (FH), and cells from the FHR1/3-/- group had somewhat higher surface-associated FH levels than those from FHR1/3+/+ individuals. FHR1/3-/- monocytes also showed elevated secretion of TNF-α, IL-1ß, and IL-10 in response to TLR7/8 or TLR4 ligands. Similarly, FHR1/3-/- mDCs and pDCs showed modest but evident hyper-responsiveness to TLR ligands. Our findings, that the FHR1/3-/- genotype is associated with significant alterations of both the relative prominence and the functioning of monocyte and DC subsets, may be relevant in understanding the mechanism underlying the association of the genotype with immune inflammatory disorders.


Asunto(s)
Proteínas Sanguíneas/genética , Proteínas Inactivadoras del Complemento C3b/genética , Genotipo , Enfermedades del Sistema Inmune/genética , Inflamación/genética , Leucocitos Mononucleares/fisiología , Eliminación de Secuencia/genética , Adulto , Células Cultivadas , Citocinas , Femenino , Homocigoto , Humanos , Inmunidad Celular , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven
6.
Eur J Immunol ; 48(8): 1285-1294, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29677388

RESUMEN

The distinct response shown by different phenotypes of macrophages and monocytes under various clinical conditions has put the heterogeneity of these cells into focus of investigation for several diseases. Recently, we have described that after engulfing hemoglobin (Hb)-activated platelets, classical monocytes differentiated into pro-inflammatory phenotypes, which were abundant in the circulation of paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease patients. Our current study shows that upon engulfment of Hb-activated platelets, monocytes differentiate into M1-macrophages under M1-polarization stimulus (GM-CSF, IFN-γ + LPS). When grown under M2-polarization stimulus (M-CSF, IL-4 + IL13), the cells exhibited an M1-like phenotype, secreted elevated levels of pro-inflammatory cytokines including TNF-α and IL-1ß, and displayed loss of the secretion of cytokine such as IL-10 and also phagocytic ability unlike the conventional M2 macrophages. Interestingly, when differentiated under the above polarization stimulus, monocytes from PNH patients expressed high levels of CD80 and phospho-STAT1, like M1 macrophages. Hemolytic mice also exhibited a gradual increase in monocyte-platelet aggregates in circulation and accumulation of CD80high macrophages in thioglycollate-induced inflamed peritoneum. The spleen of the mice was also populated by CD80high macrophages with compromised phagocytic capacity. Our findings suggest that the hemolytic environment and specifically the Hb-activated platelets, which are abundant in circulation during intravascular hemolysis, closely regulate monocyte differentiation.


Asunto(s)
Plaquetas/inmunología , Hemoglobinas/metabolismo , Hemoglobinuria Paroxística/patología , Hemólisis/inmunología , Macrófagos/citología , Monocitos/citología , Fagocitosis/inmunología , Anemia de Células Falciformes/inmunología , Anemia de Células Falciformes/patología , Animales , Antígeno B7-1/metabolismo , Modelos Animales de Enfermedad , Hemoglobinuria Paroxística/inmunología , Humanos , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Blood Cells Mol Dis ; 79: 102349, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31357175

RESUMEN

Complement factor H (FH) and FH-related proteins (FHRs), structurally similar proteins are involved in the regulation of complement activation. Homozygous deletion of FHR 1 and 3 proteins (FHR1/3-/-) is known as a risk factor for disorders such as aHUS and SLE, characterised by thrombo-inflammatory complications. Interestingly, FHR1/3-/- genotype also exists as polymorphism in healthy population of various ethnicities around the world including 8-10% Indians. In an effort to understand the functional role of this polymorphism, we describe in this study an elevated surface-bound FH on platelets and monocytes, but not other blood cells in FHR1/3 -/- healthy individuals. The FHR1/3-/- platelets displayed diminish ability to form aggregates in response to agonists in vitro. The FHR1/3-/- monocytes displayed elevated secretion of TNFα, IL1ß, IL6 and IL10 in response to TLR ligands. However, exogenous FH limits platelet aggregates formation as well as cytokine secretion in monocytes. Therefore, observations together suggest a differential regulation of platelets and monocytes by FH-FHR1/3 axis in healthy individuals. While these findings will need more detailed investigation, it is clear that the connection between FH-FHR axis and thrombo-inflammatory complications is likely to be complex in diseases including aHUS and SLE, and provide interesting new directions for future study.


Asunto(s)
Plaquetas/fisiología , Proteínas Sanguíneas/deficiencia , Monocitos/fisiología , Proteínas Sanguíneas/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/farmacología , Citocinas/metabolismo , Voluntarios Sanos , Humanos , Agregación Plaquetaria
8.
Blood Cells Mol Dis ; 77: 29-33, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30953939

RESUMEN

A young Indian female visited hospital as a suspected case of thrombotic thrombocytopenic purpura (TTP) with relapsed thrombotic complications with low platelet counts, infarct in middle cerebral artery and thrombi in microvessels. We first confirmed the deficiency of ADAMTS13 metalloprotease in this patient showing improper cleavage of vWF multimers by her plasma unlike her parents and brother. Although patient had very less ADAMTS13 antigen in plasma, but it did not appear to be the cause of deficiency of the enzyme, because her father had similarly low antigen level and he never had prothrombotic complications. While investigating the genetic change in ADAMTS13, we observed four homozygous-SNPs (g.420T>C, g.1342C>G, g.1716G>A and g.2280T>C) in exon 5, 12, 15 and 19 respectively in patient and her father unlike the heterozygous form of same SNPs in mother and brother. Further to investigate the cause of ADAMTS13 deficiency, we observed an elevated level of antibody against ADAMTS13 in patient unlike her father and other family members. Our study therefore provides the molecular approach of diagnosis of TTP in this patient and also highlights the use of such techniques in India. More importantly, study provides the clue of alternate treatment such as immunosuppressant therapy to this patient.


Asunto(s)
Proteína ADAMTS13/inmunología , Autoanticuerpos/inmunología , Púrpura Trombocitopénica Trombótica/inmunología , Púrpura Trombocitopénica Trombótica/metabolismo , Proteína ADAMTS13/antagonistas & inhibidores , Proteína ADAMTS13/sangre , Proteína ADAMTS13/genética , Autoanticuerpos/sangre , Autoantígenos/inmunología , Autoinmunidad , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , India , Masculino , Pruebas de Función Plaquetaria , Polimorfismo Genético , Polimorfismo de Nucleótido Simple , Proteolisis , Púrpura Trombocitopénica Trombótica/diagnóstico , Púrpura Trombocitopénica Trombótica/genética , Factor de von Willebrand/metabolismo
9.
Immunology ; 2018 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-29485195

RESUMEN

We previously reported that Indian paediatric patients with atypical haemolytic-uraemic syndrome (aHUS) showed high frequencies of anti-complement factor H (FH) autoantibodies that are correlated with homozygous deletion of the genes for FH-related proteins 1 and 3 (FHR1 and FHR3) (FHR1/3-/- ). We now report that Indian paediatric aHUS patients without anti-FH autoantibodies also showed modestly higher frequencies of the FHR1/3-/- genotype. Further, when we characterized epitope specificities and binding avidities of anti-FH autoantibodies in aHUS patients, most anti-FH autoantibodies were directed towards the FH cell-surface anchoring polyanionic binding site-containing C-terminal short conservative regions (SCRs) 17-20 with higher binding avidities than for native FH. FH SCR17-20-binding anti-FH autoantibodies also bound the other cell-surface anchoring polyanionic binding site-containing region FH SCR5-8, at lower binding avidities. Anti-FH autoantibody avidities correlated with antibody titres. These anti-FH autoantibody characteristics did not differ between aHUS patients with or without the FHR1/3-/- genotype. Our data suggest a complex matrix of interactions between FHR1-FHR3 deletion, immunomodulation and anti-FH autoantibodies in the aetiopathogenesis of aHUS.

10.
Clin Immunol ; 175: 133-142, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28039017

RESUMEN

Monocytes and macrophage combat infections and maintain homeostatic balance by engulfing microbes and apoptotic cells, and releasing inflammatory cytokines. Studies have described that these cells develop anti-inflammatory properties upon recycling the free-hemoglobin (Hb) in hemolytic conditions. While investigating the phenotype of monocytes in two hemolytic disorders-paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD), we observed a high number of pro-inflammatory (CD14+CD16hi) monocytes in these patients. We further investigated in vitro the phenotype of these monocytes and found an estimated 55% of CD14+ cells were transformed into the CD14+CD16hi subset after engulfing Hb-activated platelets. The CD14+CD16hi monocytes, which were positive for both intracellular Hb and CD42b (platelet marker), secreted significant amounts of TNF-α and IL-1ß, unlike monocytes treated with only free Hb, which secreted more IL-10. We have shown recently the presence of a high number of Hb-bound hyperactive platelets in patients with both diseases, and further investigated if the monocytes engulfed these activated platelets in vivo. As expected, we found 95% of CD14+CD16hi monocytes with both intracellular Hb and CD42b in both diseases, and they expressed high TNF-α. Furthermore our data showed that these monocytes whether from patients or developed in vitro after treatment with Hb-activated platelets, secreted significant amounts of tissue factor. Besides, these CD14+CD16hi monocytes displayed significantly decreased phagocytosis of E. coli. Our study therefore suggests that this alteration of monocyte phenotype may play a role in the increased propensity to pro-inflammatory/coagulant complications observed in these hemolytic disorders-PNH and SCD.


Asunto(s)
Anemia de Células Falciformes/patología , Plaquetas/patología , Hemoglobinas/metabolismo , Hemoglobinuria Paroxística/patología , Inflamación/patología , Monocitos/patología , Anemia de Células Falciformes/metabolismo , Plaquetas/metabolismo , Hemoglobinuria Paroxística/metabolismo , Humanos , Inflamación/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Monocitos/metabolismo , Fenotipo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Receptores de IgG/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Blood ; 126(20): 2338-41, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26307534

RESUMEN

Intravascular hemolysis occurs in patients on extracorporeal membrane oxygenation. High levels of free acellular adult hemoglobin (free HbA) are associated with clotting in this mechanical device that can result in thrombotic complications. Adsorption of fibrinogen onto the surface of biomaterial correlates with platelet adhesion, which is mediated by von Willebrand factor (VWF). Because free Hb interacts with VWF, we studied the effect of hemoglobin (Hb) on platelet adhesion to fibrin(ogen) under conditions of different hydrodynamic forces. This effect was investigated using purified human HbA and fibrinogen, extracellular matrix, collagen, or purified plasma VWF as surface-coated substrates to examine flow-dependent platelet adhesion. Antibodies and VWF-deficient plasma were also used. Free Hb (≥50 mg/dL) effectively augmented platelet adhesion, and microthrombi formation on fibrin(ogen), extracellular matrix, and collagen at high shear stress. The effect of free Hb was effectively blocked by anti-glycoprotein Ibα (GPIbα) antibodies or depletion of VWF. Unexpectedly, free Hb also promoted firm platelet adhesion and stable microthrombi on VWF. Lastly, we determined that Hb interacts directly with the A1 domain. This study is the first to demonstrate that extracellular Hb directly affects the GPIbα-VWF interaction in thrombosis, and describes another mechanism by which hemolysis is connected to thrombotic events.


Asunto(s)
Plaquetas/metabolismo , Hemodinámica , Hemoglobinas/metabolismo , Adhesividad Plaquetaria , Trombosis/metabolismo , Factor de von Willebrand/metabolismo , Anticuerpos/química , Plaquetas/química , Plaquetas/patología , Hemoglobinas/química , Humanos , Integrina alfa2/química , Integrina alfa2/metabolismo , Estructura Terciaria de Proteína , Trombosis/patología , Factor de von Willebrand/química
12.
IUBMB Life ; 67(7): 506-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26177573

RESUMEN

The rapid outbreak of type-2 diabetes is one of the largest public health problems around the globe. Particularly, the developing nations are becoming the epicenters of cardiometabolic disorders owing to the change in lifestyle and diet preference besides genetic predisposition. Diabetes has become a major independent risk factor for cardiovascular diseases in South Asian countries including India. The pathogenesis of type-2 diabetes primarily initiates with inadequacy of pancreatic islet ß-cells to respond to chronic fuel surfeit and hence causing glycemic load, insulin resistance, and obesity. Urban Indian life is threatened with unhealthy high calorie diet and sedentary habits, and thus impairing the metabolic status of "thin-fat Indians" and rendering them more vulnerable to metabolic disorders. Furthermore, the metabolic dysfunction may be triggered off quite early in life due to poor maternal health and impairment in intrauterine programming and, particularly in rural India. The impaired fetal development affects the health status in later stage of life by promoting obesity, insulin resistance, type-2 diabetes, and cardiovascular complications. Therefore, the preventive and therapeutic approaches focus on a holistic strategy to improve maternal and child health, promote balanced diet and physical exercise in combination with pharmacological intervention of reducing/checking hyperglycemia, obesity, and cardiovascular complications. This review summarizes the epidemiology, mechanisms, and risk factors for diabetes and cardiovascular disorders with a focus on the Indian subcontinent.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/fisiopatología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/prevención & control , Diabetes Gestacional/etiología , Dieta , Dislipidemias/etiología , Femenino , Humanos , India/epidemiología , Resistencia a la Insulina , Obesidad/etiología , Embarazo , Factores de Riesgo
13.
Haematologica ; 100(12): 1526-33, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26341739

RESUMEN

Intravascular hemolysis increases the risk of hypercoagulation and thrombosis in hemolytic disorders. Our study shows a novel mechanism by which extracellular hemoglobin directly affects platelet activation. The binding of Hb to glycoprotein1bα activates platelets. Lower concentrations of Hb (0.37-3 µM) significantly increase the phosphorylation of signaling adapter proteins, such as Lyn, PI3K, AKT, and ERK, and promote platelet aggregation in vitro. Higher concentrations of Hb (3-6 µM) activate the pro-apoptotic proteins Bak, Bax, cytochrome c, caspase-9 and caspase-3, and increase platelet clot formation. Increased plasma Hb activates platelets and promotes their apoptosis, and plays a crucial role in the pathogenesis of aggregation and development of the procoagulant state in hemolytic disorders. Furthermore, we show that in patients with paroxysmal nocturnal hemoglobinuria, a chronic hemolytic disease characterized by recurrent events of intravascular thrombosis and thromboembolism, it is the elevated plasma Hb or platelet surface bound Hb that positively correlates with platelet activation.


Asunto(s)
Apoptosis , Plaquetas/metabolismo , Hemoglobinas/metabolismo , Hemólisis , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Plaquetas/patología , Femenino , Hemoglobinuria Paroxística/patología , Humanos , Masculino
14.
FEBS J ; 291(2): 376-391, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37845743

RESUMEN

Platelet activation and related cardiovascular complications are the hallmarks of type 2 diabetes (T2D). We investigated the mechanism of platelet activation in T2D using MS-based identification of differentially expressed platelet proteins with a focus on glycosylated forms. Glycosylation is considered one of the common post-translational modifications in T2D, and N/O-linked glycosylation of glycoproteins (GPs)/integrins is known to play crucial roles in platelet activation. Our platelet proteome data revealed elevated levels of GPs GPIbα, GPIIbIIIa, GPIV (CD36), GPV and integrins in T2D patients. T2D platelets had elevated N-linked glycosylation of CD36 at asparagine (Asn)408,417 . Enrichment analysis revealed a close association of glycosylated CD36 with thrombospondin-1, fibrinogen and SERPINA1 in T2D platelets. The glycosylation of CD36 has previously been reported to increase cellular uptake of long-chain fatty acids. Our in silico molecular docking data also showed a favorable binding of cholesterol with glycosylated Asn417 CD36 compared to the non-glycosylated form. We further investigated the CD36:LDL cholesterol axis in T2D. Elevated levels of oxidized-low density lipoprotein (oxLDL) were found to cause significant platelet activation via CD36-mediated stimulation of Lyn-JNK signaling. Sulfo-N-succinimidyl oleate, an inhibitor of CD36, effectively inhibited oxLDL-mediated platelet activation and adhesion in vitro. Our study suggests increased glycosylation of CD36 in T2D platelets as a potential route for oxLDL-mediated platelet activation. The oxLDL:CD36 axis may thus be exploited as a prospective target to develop therapeutics against thrombosis in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/genética , Glicosilación , Simulación del Acoplamiento Molecular , Activación Plaquetaria/fisiología , Lipoproteínas LDL/farmacología , Factores de Riesgo , Integrinas/metabolismo
15.
J Thromb Haemost ; 22(3): 818-833, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38029855

RESUMEN

BACKGROUND: Activated platelets secrete platelet factor 4 (PF4), which contributes to viral pathogenesis. Recently, we reported the proviral role of PF4 in replication of closely related flaviviruses, Japanese encephalitis virus (JEV) and dengue virus (DENV). OBJECTIVES: This study aimed to investigate the detailed mechanism of PF4-mediated virus replication. METHODS: PF4-/- or wild-type (WT) mice were infected with JEV, and host defense mechanisms, including autophagic/interferon (IFN) responses, were assessed. WT mice were pretreated with the CXCR3 antagonist AMG487 that inhibits PF4:CXCR3 pathway. This pathway was tested in PF4-/- monocytes infected with DENV or in monocytes isolated from patients with DENV infection. RESULTS: PF4-/- mice infected with JEV showed reduced viral load and improved brain inflammation and survival. PF4-/- mice synthesized more IFN-α/ß with higher expression of phosphorylated IRF3 in the brain. PF4 treatment decreased IRF-3/7/9 and IFN-α/ß expression and suppressed autophagic LC3-II flux and lysosomal degradation of viral proteins in JEV-infected cells. PF4 increased the expression of P-mTOR, P-p38, and P-ULK1Ser757 and decreased expression of LC3-II. Decreased autophagosome-lysosome fusion in turn promoted DENV2 replication. The above processes were reversed by AMG487. Uninfected PF4-/- monocytes showed elevated LC3-II and autophagosome-lysosome fusion. Microglia of JEV-infected PF4-/- mice exhibited elevated LC3-II inversely related to viral load. Similarly, monocytes from PF4-/- mice showed reduced infection by DENV2. In patients with DENV infection, higher plasma PF4 and viral load were inversely correlated with LC3-II, LAMP-1, and lysosomal degradation of DENV-NS1 in monocytes during the febrile phase. CONCLUSION: These studies suggest that PF4 deficiency or inhibition of the PF4:CXCR3 pathway prevents JEV and DENV infection. The studies also highlight the PF4:CXCR3 axis as a potential target to develop treatment regimens against flaviviruses.


Asunto(s)
Dengue , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Pirimidinonas , Animales , Humanos , Ratones , Acetamidas , Dengue/tratamiento farmacológico , Dengue/metabolismo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/tratamiento farmacológico , Factores Inmunológicos , Factor Plaquetario 4 , Receptores CXCR3
16.
mBio ; : e0182323, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37982662

RESUMEN

IMPORTANCE: Severe dengue manifestations caused by the dengue virus are a global health problem. Studies suggest that severe dengue disease depends on uncontrolled immune cell activation, and excessive inflammation adds to the pathogenesis of severe dengue disease. Therefore, it is important to understand the process that triggers the uncontrolled activation of the immune cells. The change in immune response in mild to severe dengue may be due to direct virus-to-cell interaction or it could be a contact-independent process through the extracellular vesicles (EVs) released from infected cells. The importance of circulating EVs in the context of dengue virus infection and pathogenesis remains unexplored. Therefore, understanding the possible biological function of circulating EVs may help to delineate the role of EVs in the progression of disease. Our present study highlights that EVs from plasma of severe dengue patients can have immunosuppressive properties on CD4+ T cells which may contribute to T cell suppression and may contribute to dengue disease progression.

17.
Virus Res ; 309: 198668, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34971702

RESUMEN

Dengue virus (DENV) is most prevalent arthropod-borne human pathogen belongs to Flaviviridae family causes thousands of deaths annually. HMGB1 is highly conserved, ubiquitously expressed, non-histone nuclear protein which plays important role in diseases like metabolic disorders, cancer, and viral infections. However, the importance of HMGB1 in DENV infection is understudied. In this study, we observed that DENV-2 induces cytoplasmic translocation and secretion of HMGB1. Interestingly, inhibition of HMGB1 secretion by ethyl pyruvate (EP) enhanced viral propagation while silencing of HMGB1 resulted in abrogated viral replication in DENV-2 infected A549 cells. RNA-Electrophoretic mobility shift assay and immunoprecipitation showed that HMGB1 interacts with 5'-3' UTRs of DENV-2 genome. This interaction further stimulates production of proinflammatory cytokines like TNF-α, IL-6 and IL-1ß which have been implicated in pathogenesis of severe DENV disease. Together, our finding suggests that DENV-2 modulates HMGB1 translocation and HMGB1-DENV-2 UTRs RNA interaction further induces proinflammatory cytokines production in A549 cells. This study discloses HMGB1 as an important host factor contributing to disease pathogenesis and hence can be targeted as an alternative approach for antiviral development against DENV virus infection.


Asunto(s)
Virus del Dengue , Dengue , Proteína HMGB1 , Regiones no Traducidas 5' , Virus del Dengue/fisiología , Genoma Viral , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Replicación Viral
18.
Front Cell Dev Biol ; 10: 834016, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35386203

RESUMEN

Prion peptide (PrP) misfolds to infectious scrapie isoform, the ß pleat-rich insoluble fibrils responsible for neurodegeneration and fatal conformational diseases in humans. The amino acid sequence 106-126 from prion proteins, PrP(106-126), is highly amyloidogenic and implicated in prion-induced pathologies. Here, we report a novel interaction between PrP(106-126) and the thrombogenic plasma protein fibrinogen that can lead to mitigation of prion-mediated pro-thrombotic responses in human platelets as well as significant decline in neuronal toxicity. Thus, prior exposure to fibrinogen-restrained PrP-induced rise in cytosolic calcium, calpain activation, and shedding of extracellular vesicles in platelets while it, too, averted cytotoxicity of neuronal cells triggered by prion peptide. Interestingly, PrP was found to accelerate fibrin-rich clot formation, which was resistant to plasmin-mediated fibrinolysis, consistent with enhanced thrombus stability provoked by PrP. We propose that PrP-fibrinogen interaction can be clinically exploited further for prevention and management of infectious prion related disorders. Small molecules or peptides mimicking PrP-binding sites on fibrinogen can potentially mitigate PrP-induced cellular toxicity while also preventing the negative impact of PrP on fibrin clot formation and lysis.

19.
Biomater Adv ; 143: 213184, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36371969

RESUMEN

Herein, we report redox responsive, colon cancer targeting poly(allylamine) (PA)/eudragit S-100 (EU) nanoparticles (PAEU NPs) (≈59 nm). These disulfide crosslinked PAEU NPs are developed via air oxidation of thiolated PA and thiolated EU, eliminating the need of any external crosslinking agent for dual drug delivery. PAEU NPs can effectively encapsulate both hydrophilic doxorubicin (DOX) and hydrophobic curcumin (Cur) drug with ≈85 % and ≈97 % encapsulation efficiency respectively. Here, the combination of drugs having different anticancer mechanism offers the possibility of developing nanosystem with enhanced anticancer efficacy. The developed PAEU NPs show good colloidal stability and low drug release under physiological conditions, while high DOX (≈98 %) and Cur (≈93 %) release is observed in reducing environment (10 mM GSH). Further, DOX and Cur loaded PAEU NPs exhibit higher cancer cell killing efficiency as compared to individual free drugs. In vivo biodistribution studies with Balb/C mice display the retention of PAEU NPs in the colon region up to 24 h presenting the developed approach as an efficient way for colorectal cancer therapy.


Asunto(s)
Alilamina , Neoplasias Colorrectales , Curcumina , Nanopartículas , Ratones , Animales , Distribución Tisular , Doxorrubicina/uso terapéutico , Oxidación-Reducción , Neoplasias Colorrectales/tratamiento farmacológico
20.
Biomater Adv ; 136: 212796, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35929295

RESUMEN

Inflammatory Bowel (IBD) is an umbrella term which includes Crohn's Disease (CD) and Ulcerative Colitis (UC). At present, therapies available for management of the UC includes, corticosteroid, immuno-suppressants and antibiotics are used for mild to moderate UC conditions which can cause nephrotoxicity, hepatotoxicity and cardiotoxicity. Hence, a novel therapeutic candidate having potent anti-inflammatory effect is urgently warranted for the management of UC. Melatonin has emerged as a potent anti-inflammatory agent. However, poor solubility limits its therapeutic potential. Therefore, colon targeted Eudragit-S-100 coated chitosan nanoparticles have been demonstrated to improve melatonin therapeutic efficacy. It was found that melatonin loaded chitosan and colon targeted chitosan nanoparticles had promising anti-inflammatory efficacy in terms of NO scavenging activity in an in-vitro LPS challenged macrophages. Also, colon targeted oral chitosan nano-formulation exhibited remarkable protection in an in vivo UC mice model by improving gross pathological parameters, histo-architectural protection, goblet cell depletion, and immune cells infiltration which can be extrapolated to clinical studies.


Asunto(s)
Quitosano , Colitis Ulcerosa , Enfermedades Inflamatorias del Intestino , Melatonina , Animales , Antiinflamatorios/farmacología , Quitosano/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inducido químicamente , Melatonina/farmacología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA