Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 141(4): 406-421, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36395340

RESUMEN

Glycosylation is recognized as a key process for proper megakaryopoiesis and platelet formation. The enzyme uridine diphosphate (UDP)-galactose-4-epimerase, encoded by GALE, is involved in galactose metabolism and protein glycosylation. Here, we studied 3 patients from 2 unrelated families who showed lifelong severe thrombocytopenia, bleeding diathesis, mental retardation, mitral valve prolapse, and jaundice. Whole-exome sequencing revealed 4 variants that affect GALE, 3 of those previously unreported (Pedigree A, p.Lys78ValfsX32 and p.Thr150Met; Pedigree B, p.Val128Met; and p.Leu223Pro). Platelet phenotype analysis showed giant and/or grey platelets, impaired platelet aggregation, and severely reduced alpha and dense granule secretion. Enzymatic activity of the UDP-galactose-4-epimerase enzyme was severely decreased in all patients. Immunoblotting of platelet lysates revealed reduced GALE protein levels, a significant decrease in N-acetyl-lactosamine (LacNAc), showing a hypoglycosylation pattern, reduced surface expression of gylcoprotein Ibα-IX-V (GPIbα-IX-V) complex and mature ß1 integrin, and increased apoptosis. In vitro studies performed with patients-derived megakaryocytes showed normal ploidy and maturation but decreased proplatelet formation because of the impaired glycosylation of the GPIbα and ß1 integrin, and reduced externalization to megakaryocyte and platelet membranes. Altered distribution of filamin A and actin and delocalization of the von Willebrand factor were also shown. Overall, this study expands our knowledge of GALE-related thrombocytopenia and emphasizes the critical role of GALE in the physiological glycosylation of key proteins involved in platelet production and function.


Asunto(s)
Trombocitopenia , UDPglucosa 4-Epimerasa , Humanos , Plaquetas/metabolismo , Galactosa/metabolismo , Glicosilación , Integrina beta1/metabolismo , Megacariocitos/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombopoyesis/genética , UDPglucosa 4-Epimerasa/genética , UDPglucosa 4-Epimerasa/metabolismo , Uridina Difosfato/metabolismo
2.
Am J Hematol ; 99(3): 336-349, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38165047

RESUMEN

Mechanisms through which mature megakaryocytes (Mks) and their progenitors sense the bone marrow extracellular matrix to promote lineage differentiation in health and disease are still partially understood. We found PIEZO1, a mechanosensitive cation channel, to be expressed in mouse and human Mks. Human mutations in PIEZO1 have been described to be associated with blood cell disorders. Yet, a role for PIEZO1 in megakaryopoiesis and proplatelet formation has never been investigated. Here, we show that activation of PIEZO1 increases the number of immature Mks in mice, while the number of mature Mks and Mk ploidy level are reduced. Piezo1/2 knockout mice show an increase in Mk size and platelet count, both at basal state and upon marrow regeneration. Similarly, in human samples, PIEZO1 is expressed during megakaryopoiesis. Its activation reduces Mk size, ploidy, maturation, and proplatelet extension. Resulting effects of PIEZO1 activation on Mks resemble the profile in Primary Myelofibrosis (PMF). Intriguingly, Mks derived from Jak2V617F PMF mice show significantly elevated PIEZO1 expression, compared to wild-type controls. Accordingly, Mks isolated from bone marrow aspirates of JAK2V617F PMF patients show increased PIEZO1 expression compared to Essential Thrombocythemia. Most importantly, PIEZO1 expression in bone marrow Mks is inversely correlated with patient platelet count. The ploidy, maturation, and proplatelet formation of Mks from JAK2V617F PMF patients are rescued upon PIEZO1 inhibition. Together, our data suggest that PIEZO1 places a brake on Mk maturation and platelet formation in physiology, and its upregulation in PMF Mks might contribute to aggravating some hallmarks of the disease.


Asunto(s)
Mielofibrosis Primaria , Trombocitemia Esencial , Humanos , Animales , Ratones , Megacariocitos/metabolismo , Mielofibrosis Primaria/genética , Médula Ósea , Trombopoyesis/genética , Trombocitemia Esencial/metabolismo , Plaquetas/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo
3.
Blood ; 137(15): 2085-2089, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33238000

RESUMEN

Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure ß4-N-acetyllactosamine (Galß1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding ß-1,4-galactosyltransferase 1 (ß4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify ß4GalT1 as a potential actionable target for therapy.


Asunto(s)
Plaquetas/patología , Galactosa/metabolismo , Galactosiltransferasas/genética , Trastornos Mieloproliferativos/genética , Trombopoyetina/sangre , Plaquetas/metabolismo , Galactosa/análisis , Galactosiltransferasas/metabolismo , Humanos , Megacariocitos/metabolismo , Megacariocitos/patología , Mutación , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/metabolismo , Trombopoyetina/metabolismo , Regulación hacia Arriba
4.
Haematologica ; 108(4): 1141-1157, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36546455

RESUMEN

Cardiovascular (CV) disease prevention with low-dose aspirin can be less effective in patients with a faster recovery of platelet (PLT) cyclooxygenase (COX)-1 activity during the 24-hour dosing interval. We previously showed that incomplete suppression of TXA2 over 24 hours can be rescued by a twice daily aspirin regimen. Here we show that reduced PLT glycoprotein (GP)Ibα shedding characterizes patients with accelerated COX-1 recovery and may contribute to higher thrombopoietin (TPO) production and higher rates of newly formed PLT, escaping aspirin inhibition over 24 hours. Two hundred aspirin-treated patients with high CV risk (100 with type 2 diabetes mellitus) were stratified according to the kinetics of PLT COX-1 activity recovery during the 10- to 24-hour dosing interval. Whole proteome analysis showed that PLT from patients with accelerated COX-1 recovery were enriched in proteins involved in cell survival, inhibition of apoptosis and cellular protrusion formation. In agreement, we documented increased plasma TPO, megakaryocyte maturation and proplatelet formation, and conversely increased PLT galactose and reduced caspase 3, phosphatidylserine exposure and ADAM17 activation, translating into diminished GPIbα cleavage and glycocalicin (GC) release. Treatment of HepG2 cells with recombinant GC led to a dose-dependent reduction of TPO mRNA in the liver, suggesting that reduced GPIbα ectodomain shedding may unleash thrombopoiesis. A cluster of clinical markers, including younger age, non-alcoholic fatty liver disease, visceral obesity and higher TPO/GC ratio, predicted with significant accuracy the likelihood of faster COX-1 recovery and suboptimal aspirin response. Circulating TPO/GC ratio, reflecting a dysregulation of PLT lifespan and production, may provide a simple tool to identify patients amenable to more frequent aspirin daily dosing.


Asunto(s)
Diabetes Mellitus Tipo 2 , Trombocitopenia , Humanos , Aspirina/farmacología , Trombopoyesis , Diabetes Mellitus Tipo 2/metabolismo , Plaquetas/metabolismo , Trombocitopenia/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo
5.
Blood ; 135(2): 133-144, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31697806

RESUMEN

Approximately one-fourth of patients with essential thrombocythemia or primary myelofibrosis carry a somatic mutation of the calreticulin gene (CALR), the gene encoding for calreticulin. A 52-bp deletion (type I mutation) and a 5-bp insertion (type II mutation) are the most frequent genetic lesions. The mechanism(s) by which a CALR mutation leads to a myeloproliferative phenotype has been clarified only in part. We studied the interaction between calreticulin and store-operated calcium (Ca2+) entry (SOCE) machinery in megakaryocytes (Mks) from healthy individuals and from patients with CALR-mutated myeloproliferative neoplasms (MPNs). In Mks from healthy subjects, binding of recombinant human thrombopoietin to c-Mpl induced the activation of signal transducer and activator of transcription 5, AKT, and extracellular signal-regulated kinase 1/2, determining inositol triphosphate-dependent Ca2+ release from the endoplasmic reticulum (ER). This resulted in the dissociation of the ER protein 57 (ERp57)-mediated complex between calreticulin and stromal interaction molecule 1 (STIM1), a protein of the SOCE machinery that leads to Ca2+ mobilization. In Mks from patients with CALR-mutated MPNs, defective interactions between mutant calreticulin, ERp57, and STIM1 activated SOCE and generated spontaneous cytosolic Ca2+ flows. In turn, this resulted in abnormal Mk proliferation that was reverted using a specific SOCE inhibitor. In summary, the abnormal SOCE regulation of Ca2+ flows in Mks contributes to the pathophysiology of CALR-mutated MPNs. In perspective, SOCE may represent a new therapeutic target to counteract Mk proliferation and its clinical consequences in MPNs.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Calreticulina/genética , Calreticulina/metabolismo , Megacariocitos/patología , Mutación , Trastornos Mieloproliferativos/patología , Canales de Calcio Activados por la Liberación de Calcio/genética , Estudios de Casos y Controles , Humanos , Megacariocitos/metabolismo , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
6.
Platelets ; 31(6): 717-723, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32522064

RESUMEN

Mechanisms related to platelet release in the context of the bone marrow niche are not completely known. In this review we discuss what has been discovered about four critical aspects of this process: 1) the bone marrow niche organization, 2) the role of the extracellular matrix components, 3) the mechanisms by which megakaryocytes release platelets and 4) the novel approaches to mimic the bone marrow environment and produce platelets ex vivo.


Asunto(s)
Plaquetas/metabolismo , Animales , Humanos
7.
Am J Hematol ; 93(5): 615-622, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29359451

RESUMEN

Splenic hematopoiesis is a major feature in the course of myelofibrosis (MF). In fact, the spleen of patients with MF contains malignant hematopoietic stem cells retaining a complete differentiation program, suggesting both a pivotal role of the spleen in maintaining the disease and a tight regulation of hematopoiesis by the splenic microenvironment, in particular by mesenchymal stromal cells (MSCs). Little is known about splenic MSCs (Sp-MSCs), both in normal and in pathological context. In this work, we have in vitro expanded and characterized Sp-MSCs from 25 patients with MF and 13 healthy subjects (HS). They shared similar phenotype, growth kinetics, and differentiation capacity. However, MF Sp-MSCs expressed significant lower levels of nestin, and favored megakaryocyte (Mk) differentiation in vitro at a larger extent than their normal counterpart. Moreover, they showed a significant upregulation of matrix metalloprotease 2 (MMP2) and fibronectin 1 (FN1) genes both at mRNA expression and at protein level, and, finally, developed genetic abnormalities which were never detected in HS-derived Sp-MSCs. Our data point toward the existence of a defective splenic niche in patients with MF that could be responsible of some pathological features of the disease, including the increased trafficking of CD34+ cells and the expansion of the megakaryocytic lineage.


Asunto(s)
Células Madre Mesenquimatosas/patología , Mielofibrosis Primaria/patología , Bazo/patología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD34 , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Femenino , Fibronectinas/metabolismo , Hematopoyesis , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Megacariocitos/patología , Persona de Mediana Edad , Nestina/metabolismo , Adulto Joven
8.
J Biol Chem ; 291(9): 4343-55, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26702058

RESUMEN

A bacterial collagen-like protein Scl2 has been developed as a recombinant collagen model system to host human collagen ligand-binding sequences, with the goal of generating biomaterials with selective collagen bioactivities. Defined binding sites in human collagen for integrins, fibronectin, heparin, and MMP-1 have been introduced into the triple-helical domain of the bacterial collagen and led to the expected biological activities. The modular insertion of activities is extended here to the discoidin domain receptors (DDRs), which are collagen-activated receptor tyrosine kinases. Insertion of the DDR-binding sequence from human collagen III into bacterial collagen led to specific receptor binding. However, even at the highest testable concentrations, the construct was unable to stimulate DDR autophosphorylation. The recombinant collagen expressed in Escherichia coli does not contain hydroxyproline (Hyp), and complementary synthetic peptide studies showed that replacement of Hyp by Pro at the critical Gly-Val-Met-Gly-Phe-Hyp position decreased the DDR-binding affinity and consequently required a higher concentration for the induction of receptor activation. The ability of the recombinant bacterial collagen to bind the DDRs without inducing kinase activation suggested it could interfere with the interactions between animal collagen and the DDRs, and such an inhibitory role was confirmed in vitro and with a cell migration assay. This study illustrates that recombinant collagen can complement synthetic peptides in investigating structure-activity relationships, and this system has the potential for the introduction or inhibition of specific biological activities.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colágeno Tipo III/metabolismo , Colágeno/metabolismo , Megacariocitos/metabolismo , Modelos Moleculares , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Movimiento Celular , Células Cultivadas , Colágeno/química , Colágeno/genética , Colágeno Tipo III/química , Colágeno Tipo III/genética , Receptores con Dominio Discoidina , Sangre Fetal/citología , Células HEK293 , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Ligandos , Megacariocitos/citología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/química , Receptores Mitogénicos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Streptococcus pyogenes
9.
Stem Cells ; 34(4): 1123-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26748484

RESUMEN

Extracellular matrix (ECM) components initiate crucial biochemical and biomechanical cues that are required for bone marrow homeostasis. In our research, we prove that a peri-cellular matrix composed primarily of type III and type IV collagens, and fibronectin surrounds human megakaryocytes in the bone marrow. The data we collected support the hypothesis that bone marrow megakaryocytes possess a complete mechanism to synthesize the ECM components, and that thrombopoietin is a pivotal regulator of this new function inducing transforming growth factor-ß1 (TGF-ß1) release and consequent activation of the downstream pathways, both in vitro and in vivo. This activation results in a dose dependent increase of ECM component synthesis by megakaryocytes, which is reverted upon incubation with JAK and TGF-ß1 receptor specific inhibitors. These data are pivotal for understanding the central role of megakaryocytes in creating their own regulatory niche within the bone marrow environment.


Asunto(s)
Matriz Extracelular/metabolismo , Megacariocitos/metabolismo , Trombopoyetina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Médula Ósea/crecimiento & desarrollo , Médula Ósea/metabolismo , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Matriz Extracelular/genética , Sangre Fetal/citología , Sangre Fetal/metabolismo , Fibronectinas/metabolismo , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Megacariocitos/efectos de los fármacos , Ratones , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Trombopoyetina/genética , Factor de Crecimiento Transformador beta1/genética
10.
Haematologica ; 102(7): 1150-1160, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28411253

RESUMEN

Megakaryocytes (MK) in the bone marrow (BM) are immersed in a network of extracellular matrix components that regulates platelet release into the circulation. Combining biological and bioengineering approaches, we found that the activation of transient receptor potential cation channel subfamily V member 4 (TRPV4), a mechano-sensitive ion channel, is induced upon MK adhesion on softer matrices. This response promoted platelet production by triggering a cascade of events that lead to calcium influx, ß1 integrin activation and internalization, and Akt phosphorylation, responses not found on stiffer matrices. Lysyl oxidase (LOX) is a physiological modulator of BM matrix stiffness via collagen crosslinking. In vivo inhibition of LOX and consequent matrix softening lead to TRPV4 activation cascade and increased platelet levels. At the same time, in vitro proplatelet formation was reduced on a recombinant enzyme-mediated stiffer collagen. These results suggest a novel mechanism by which MKs, through TRPV4, sense extracellular matrix environmental rigidity and release platelets accordingly.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Trombopoyesis , Animales , Calcio/metabolismo , Adhesión Celular , Diferenciación Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Integrina beta1/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas , Proteína-Lisina 6-Oxidasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales Catiónicos TRPV/metabolismo
11.
Exp Cell Res ; 346(1): 1-8, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-26027944

RESUMEN

Hyaluronan (HA) is a glycosamminoglican involved in cell biology as well as a relevant polymer for tissue engineering and regenerative medicine. Megakaryocytes (Mks) are immersed in a mesh of extracellular matrix (ECM) components that regulate their maturation in the bone marrow (BM) and the release of platelets into the bloodstream. While fibrous ECMs such as collagens and fibronectin have been demonstrated to differently regulate Mk function and platelet release, the role of HA, that fills the majority of the BM extracellular interstitial space, has not been investigated so far. Here we demonstrated that, although human Mks express HA receptors, they are not affected by HA in terms of in vitro differentiation, maturation and platelet formation. Importantly, chemical properties of HA were exploited to generate hydrogels with entrapped ECMs that represent a useful model to more closely mimic the tridimensional characteristics of the BM environment for studying Mk function. In conclusion, in this work we demonstrated that HA is an ideal candidate for a 3D ex vivo model of human BM ECM component environment.


Asunto(s)
Uniones Célula-Matriz/metabolismo , Matriz Extracelular/metabolismo , Ácido Hialurónico/farmacología , Hidrogeles/farmacología , Megacariocitos/citología , Modelos Biológicos , Diferenciación Celular/efectos de los fármacos , Uniones Célula-Matriz/efectos de los fármacos , Células Cultivadas , Glucuronosiltransferasa/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Imagenología Tridimensional , Isoenzimas/metabolismo , Megacariocitos/efectos de los fármacos , Megacariocitos/enzimología , Peso Molecular , Trombopoyesis/efectos de los fármacos , Andamios del Tejido/química
12.
Cell Mol Life Sci ; 72(8): 1517-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25572292

RESUMEN

Megakaryocytes are rare cells found in the bone marrow, responsible for the everyday production and release of millions of platelets into the bloodstream. Since the discovery and cloning, in 1994, of their principal humoral factor, thrombopoietin, and its receptor c-Mpl, many efforts have been directed to define the mechanisms underlying an efficient platelet production. However, more recently different studies have pointed out new roles for megakaryocytes as regulators of bone marrow homeostasis and physiology. In this review we discuss the interaction and the reciprocal regulation of megakaryocytes with the different cellular and extracellular components of the bone marrow environment. Finally, we provide evidence that these processes may concur to the reconstitution of the bone marrow environment after injury and their deregulation may lead to the development of a series of inherited or acquired pathologies.


Asunto(s)
Médula Ósea/metabolismo , Megacariocitos/metabolismo , Animales , Plaquetas/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Matriz Extracelular/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Trombocitopenia/metabolismo , Trombocitopenia/patología , Trombopoyetina/metabolismo
13.
Mediators Inflamm ; 2016: 8512417, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27555670

RESUMEN

CXCL8 displays several tumor-promoting effects. Targeting and/or lowering CXCL8 concentrations within the tumor microenvironment would produce a therapeutic benefit. Aim of this study was to test the effect of IFNγ on the basal and TNFα-stimulated secretion of CXCL8 in TCP-1 and BCPAP thyroid cancer cell lines (harboring RET/PTC rearrangement and BRAF V600e mutation, resp.). Cells were incubated with IFNγ (1, 10, 100, and 1000 U/mL) alone or in combination with TNF-α (10 ng/mL) for 24 hours. CXCL8 and CXCL10 concentrations were measured in the cell supernatants. IFNγ inhibited in a dose-dependent and significant manner both the basal (ANOVA F: 22.759; p < 0.00001) and the TNFα-stimulated (ANOVA F: 15.309; p < 0.00001) CXCL8 secretions in BCPAP but not in TPC-1 cells (NS). On the other hand, IFNγ and IFNγ + TNF-α induced a significant secretion of CXCL10 in both BCPAP (p < 0.05) and TPC-1 (p < 0.05) cells. Transwell migration assay showed that (i) CXCL8 increased cell migration in both TPC-1 and BCPAP cells; (ii) IFNγ significantly reduced the migration only of BCPAP cells; and (iii) CXCL8 reverted the effect of IFNγ. These results constitute the first demonstration that IFNγ inhibits CXCL8 secretion and in turn the migration of a BRAF V600e mutated thyroid cell line.


Asunto(s)
Reordenamiento Génico , Interferón gamma/farmacología , Interleucina-8/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular Tumoral , Movimiento Celular , Quimiocina CXCL10/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Neoplasias de la Tiroides/genética , Cicatrización de Heridas
14.
J Biol Chem ; 289(8): 4941-51, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24375478

RESUMEN

Interaction of collagen with fibronectin is important for extracellular matrix assembly and regulation of cellular processes. A fibronectin-binding region in collagen was identified using unfolded fragments, but it is not clear if the native protein binds fibronectin with the same primary sequence. A recombinant bacterial collagen is utilized to characterize the sequence requirement for fibronectin binding. Chimeric collagens were generated by inserting the putative fibronectin-binding region from human collagen into the bacterial collagen sequence. Insertion of a sufficient length of human sequence conferred fibronectin affinity. The minimum sequence requirement was identified as a 6-triplet sequence near the unique collagenase cleavage site and was the same in both triple-helix and denatured states. Denaturation of the chimeric collagen increased its affinity for fibronectin, as seen for mammalian collagens. The fibronectin binding recombinant collagen did not contain hydroxyproline, indicating hydroxyproline is not essential for binding. However, its absence may account, in part, for the higher affinity of the native chimeric protein and the lower affinity of the denatured protein compared with type II collagen. Megakaryocytes cultured on chimeric collagen with fibronectin affinity showed improved adhesion and differentiation, suggesting a strategy for generating bioactive materials in biomedical applications.


Asunto(s)
Colágeno Tipo II/metabolismo , Fibronectinas/metabolismo , Desnaturalización Proteica , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Bovinos , Adhesión Celular , Proliferación Celular , Colágeno Tipo II/química , Electroforesis en Gel de Poliacrilamida , Fibronectinas/química , Humanos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Proteínas Recombinantes/química , Temperatura
15.
J Biol Chem ; 288(23): 16738-16746, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23530036

RESUMEN

Growing evidence demonstrates that extracellular matrices regulate many aspects of megakaryocyte (MK) development; however, among the different extracellular matrix receptors, integrin α2ß1 and glycoprotein VI are the only collagen receptors studied in platelets and MKs. In this study, we demonstrate the expression of the novel collagen receptor discoidin domain receptor 1 (DDR1) by human MKs at both mRNA and protein levels and provide evidence of DDR1 involvement in the regulation of MK motility on type I collagen through a mechanism based on the activity of SHP1 phosphatase and spleen tyrosine kinase (Syk). Specifically, we demonstrated that inhibition of DDR1 binding to type I collagen, preserving the engagement of the other collagen receptors, glycoprotein VI, α2ß1, and LAIR-1, determines a decrease in MK migration due to the reduction in SHP1 phosphatase activity and consequent increase in the phosphorylation level of its main substrate Syk. Consistently, inhibition of Syk activity restored MK migration on type I collagen. In conclusion, we report the expression and function of a novel collagen receptor on human MKs, and we point out that an increasing level of complexity is necessary to better understand MK-collagen interactions in the bone marrow environment.


Asunto(s)
Movimiento Celular/fisiología , Colágeno Tipo I/metabolismo , Megacariocitos/enzimología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Colágeno Tipo I/genética , Receptor con Dominio Discoidina 1 , Femenino , Humanos , Integrina alfa2beta1/genética , Integrina alfa2beta1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Megacariocitos/citología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Quinasa Syk
17.
Nanomedicine ; 10(3): 589-97, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24161383

RESUMEN

Boron neutron capture therapy (BNCT) is a radiotherapy treatment based on the accumulation in the tumor of a (10)B-containing drug and subsequent irradiation with low energy neutrons, which bring about the decay of (10)B to (7)Li and an α particle, causing the death of the neoplastic cell. The effectiveness of BNCT is limited by the low delivery and accumulation of the used boron-containing compounds. Here we report the development and the characterization of BPO4 nanoparticles (NPs) as a novel possible alternative drug for BNCT. An extensive analysis of BPO4 NP biocompatibility was performed using both mature blood cells (erythrocytes, neutrophils and platelets) and a model of hematopoietic progenitor cells. A time- and concentration-dependent cytotoxicity study was performed on neoplastic coloncarcinoma and osteosarcoma cell lines. BPO4 functionalization with folic acid, introduced to improve the uptake by tumor cells, appeared to effectively limit the unwanted effects of NPs on the analyzed blood components. FROM THE CLINICAL EDITOR: Boron neutron capture therapy (BNCT) is a radiotherapy treatment modality based on the accumulation of a (10)B-containing drug and subsequent irradiation with low energy neutrons, inducing the decay of (10)B to (7)Li and an α particle, causing neoplastic cell death. This team of authors reports on a folic acid functionalized BPO4 nanoparticle with improved characteristics compared with conventional BNCT approaches, as demonstrated in tumor cell lines, and hopefully to be followed by translational human studies.


Asunto(s)
Compuestos de Boro/farmacología , Terapia por Captura de Neutrón de Boro , Nanopartículas/química , Neoplasias/radioterapia , Fosfatos/farmacología , Compuestos de Boro/química , Compuestos de Boro/farmacocinética , Terapia por Captura de Neutrón de Boro/métodos , Línea Celular Tumoral , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Nanopartículas/metabolismo , Fosfatos/química , Fosfatos/farmacocinética
18.
Sci Rep ; 14(1): 5941, 2024 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467734

RESUMEN

Prolonged febrile seizures (FS) in children are linked to the development of temporal lobe epilepsy (MTLE). The association between these two pathologies may be ascribed to the long-term effects that FS exert on neural stem cells, negatively affecting the generation of new neurons. Among the insults associated with FS, oxidative stress is noteworthy. Here, we investigated the consequences of exposure to hydrogen peroxide (H2O2) in an induced pluripotent stem cell-derived neural stem cells (iNSCs) model of a patient affected by FS and MTLE. In our study, we compare the findings from the MTLE patient with those derived from iNSCs of a sibling exhibiting a milder phenotype defined only by FS, as well as a healthy individual. In response to H2O2 treatment, iNSCs derived from MTLE patients demonstrated an elevated production of reactive oxygen species and increased apoptosis, despite the higher expression levels of antioxidant genes and proteins compared to other cell lines analysed. Among the potential causative mechanisms of enhanced vulnerability of MTLE patient iNSCs to oxidative stress, we found that these cells express low levels of the heat shock protein HSPB1 and of the autophagy adaptor SQSTM1/p62. Pre-treatment of diseased iNSCs with the antioxidant molecule ascorbic acid restored HSBP1 and p62 expression and simultaneously reduced the levels of ROS and apoptosis. Our findings suggest the potential for rescuing the impaired oxidative stress response in diseased iNSCs through antioxidant treatment, offering a promising mechanism to prevent FS degeneration in MTLE.


Asunto(s)
Epilepsia del Lóbulo Temporal , Convulsiones Febriles , Niño , Humanos , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/metabolismo , Convulsiones Febriles/tratamiento farmacológico , Convulsiones Febriles/genética , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Ácido Ascórbico/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antioxidantes/metabolismo , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Hipocampo/metabolismo , Proteínas de Choque Térmico/metabolismo
19.
Leukemia ; 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39025985

RESUMEN

Despite increased understanding of the genomic landscape of Myeloproliferative Neoplasms (MPNs), the pathological mechanisms underlying abnormal megakaryocyte (Mk)-stromal crosstalk and fibrotic progression in MPNs remain unclear. We conducted mass spectrometry-based proteomics on mice with Romiplostim-dependent myelofibrosis to reveal alterations in signaling pathways and protein changes in Mks, platelets, and bone marrow (BM) cells. The chemokine Platelet Factor 4 (PF4)/Cxcl4 was up-regulated in all proteomes and increased in plasma and BM fluids of fibrotic mice. High TPO concentrations sustained in vitro PF4 synthesis and secretion in cultured Mks, while Ruxolitinib restrains the abnormal PF4 expression in vivo. We discovered that PF4 is rapidly internalized by stromal cells through surface glycosaminoglycans (GAGs) to promote myofibroblast differentiation. Cxcl4 gene silencing in Mks mitigated the profibrotic phenotype of stromal cells in TPO-saturated co-culture conditions. Consistently, extensive stromal PF4 uptake and altered GAGs deposition were detected in Romiplostim-treated, JAK2V617F mice and BM biopsies of MPN patients. BM PF4 levels and Mk/platelet CXCL4 expression were elevated in patients, exclusively in overt fibrosis. Finally, pharmacological inhibition of GAGs ameliorated in vivo fibrosis in Romiplostim-treated mice. Thus, our findings highlight the critical role of PF4 in the fibrosis progression of MPNs and substantiate the potential therapeutic strategy of neutralizing PF4-GAGs interaction.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA