Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Toxins (Basel) ; 15(12)2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38133194

RESUMEN

Typical hemolytic uremic syndrome (HUS) is mainly caused by Shiga toxin-producing Escherichia coli (STEC) releasing Shiga toxin 2 (Stx2). Two different structures of this AB5 toxin have been described: uncleaved, with intact B and A chains, and cleaved, with intact B and a nicked A chain consisting of two fragments, A1 and A2, connected by a disulfide bond. Despite having the same toxic effect on sensitive cells, the two forms differ in their binding properties for circulating cells, serum components and complement factors, thus contributing to the pathogenesis of HUS differently. The outcome of STEC infections and the development of HUS could be influenced by the relative amounts of uncleaved or cleaved Stx2 circulating in patients' blood. Cleaved Stx2 was identified and quantified for the first time in four out of eight STEC-infected patients' sera by a method based on the inhibition of cell-free translation. Cleaved Stx2 was present in the sera of patients with toxins bound to neutrophils and in two out of three patients developing HUS, suggesting its involvement in HUS pathogenesis, although in association with other bacterial or host factors.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli Shiga-Toxigénica , Humanos , Toxina Shiga II , Toxina Shiga , Neutrófilos , Bacterias , Infecciones por Escherichia coli/microbiología
2.
Front Immunol ; 13: 1058739, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713433

RESUMEN

Graft versus host disease (GVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (HSCT). Rabbit anti-T lymphocyte globulin (ATLG) in addition to calcineurin inhibitors and antimetabolites is a suitable strategy to prevent GVHD in several transplant settings. Randomized studies already demonstrated its efficacy in terms of GVHD prevention, although the effect on relapse remains the major concern for a wider use. Tailoring of ATLG dose on host characteristics is expected to minimize its side effects (immunological reconstitution, relapse, and infections). Here, day -6 to day +15 pharmacokinetics of active ATLG serum level was first assayed in an explorative cohort of 23 patients by testing the ability of the polyclonal serum to bind antigens on human leukocytes. Significantly lower levels of serum active ATLG were found in the patients who developed GVHD (ATLG_AUCCD45: 241.52 ± 152.16 vs. 766.63 +/- 283.52 (µg*day)/ml, p = 1.46e-5). Consistent results were obtained when the ATLG binding capacity was assessed on CD3+ and CD3+/CD4+ T lymphocytes (ATLG_AUCCD3: 335.83 ± 208.15 vs. 903.54 ± 378.78 (µg*day)/ml, p = 1.92e-4; ATLG_AUCCD4: 317.75 ± 170.70 vs. 910.54 ± 353.35 (µg*day)/ml, p = 3.78e-5. Concomitantly, at pre-infusion time points, increased concentrations of CD69+ extracellular vesicles (EVs) were found in patients who developed GVHD (mean fold 9.01 ± 1.33; p = 2.12e-5). Consistent results were obtained in a validation cohort of 12 additional ATLG-treated HSCT patients. Serum CD69+ EVs were mainly represented in the nano (i.e. 100 nm in diameter) EV compartment and expressed the leukocyte marker CD45, the EV markers CD9 and CD63, and CD103, a marker of tissue-resident memory T cells. The latter are expected to set up a host pro-inflammatory cell compartment that can survive in the recipient for years after conditioning regimen and contribute to GVHD pathogenesis. In summary, high levels of CD69+ EVs are significantly correlated with an increased risk of GVHD, and they may be proposed as a tool to tailor ATLG dose for personalized GVHD prevention.


Asunto(s)
Vesículas Extracelulares , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Humanos , Conejos , Suero Antilinfocítico/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Anticuerpos/uso terapéutico , Linfocitos , Recurrencia
3.
Res Pract Thromb Haemost ; 5(8): e12606, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34938937

RESUMEN

Recently, treatment of immune-mediated thrombotic thrombocytopenic purpura (ITTP) has changed with the advent of caplacizumab in clinical practice. The International Working Group (IWG) has recently integrated the ADAMTS-13 activity/autoantibody monitoring in consensus outcome definitions. We report three ITTP cases during the coronavirus disease 2019 pandemic, that received a systematic evaluation of ADAMTS-13 activity and autoantibodies. We describe how the introduction of caplacizumab and ADAMTS-13 monitoring could change the management of ITTP patients and discuss whether therapeutic choices should be based on the clinical response alone. ADAMTS-13 activity/antibodies were assessed every 5 days. Responses were evaluated according to updated IWG outcome definitions. These kinetics, rather than clinical remission, guided the therapy, allowing early and safe caplacizumab discontinuation and sensible administration of rituximab. Caplacizumab was cautiously discontinued after achieving ADAMTS-13 complete remission. These cases illustrate that prospective ADAMTS-13 evaluation and use of updated IWG definitions may improve real-life patients' management in the caplacizumab era.

4.
Front Oncol ; 11: 715217, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34900671

RESUMEN

Polycythemia Vera (PV) is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. Chronic inflammation is commonly observed in myeloproliferative neoplasms including PV. The inflammatory network includes the extracellular vesicles (EVs), which play a role in cell-cell communication. Recent evidence points to circulating microbial components/microbes as potential players in hemopoiesis regulation. To address the role of EVs in PV, here we investigated phenotype and microbial DNA cargo of circulating EVs through multidimensional analysis. Peripheral blood and feces were collected from PV patients (n=38) and healthy donors (n=30). Circulating megakaryocyte (MK)- and platelet (PLT)-derived EVs were analyzed by flow cytometry. After microbial DNA extraction from feces and isolated EVs, the 16S rDNA V3-V4 region was sequenced. We found that the proportion of circulating MK-derived EVs was significantly decreased in PV patients as compared with the healthy donors. By contrast, the proportion of the PLT-derived EVs was increased. Interestingly, PV was also associated with a microbial DNA signature of the isolated EVs with higher diversity and distinct microbial composition than the healthy counterparts. Of note, increased proportion of isolated lipopolysaccharide-associated EVs has been demonstrated in PV patients. Conversely, the gut microbiome profile failed to identify a distinct layout between PV patients and healthy donors. In conclusion, PV is associated with circulating EVs harbouring abnormal phenotype and dysbiosis signature with a potential role in the (inflammatory) pathogenesis of the disease.

5.
Cancers (Basel) ; 13(19)2021 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-34638452

RESUMEN

Polycythemia vera is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Circulating extracellular vesicles (EVs) are mostly of megakaryocyte (MK-EVs) and platelet (PLT-EVs) origin and, along with phosphatidylethanolamine (PE)-EVs, play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we investigated phenotype and microbial DNA cargo of EVs after isolation from the plasma of 38 patients with polycythemia vera. Increased proportion of MK-EVs and reduced proportion of PLT-EVs identify patients with thrombosis history. Interestingly, EVs from patients with thrombosis history were depleted in Staphylococcus DNA but enriched in DNA from Actinobacteria members as well as Anaerococcus. In addition, patients with thrombosis history had also lower levels of lipopolysaccharide-associated EVs. In regard to fibrosis, along with increased proportion of PE-EVs, the EVs of patients with marrow fibrosis were enriched in DNA from Collinsella and Flavobacterium. Here, we identified a polycythemia-vera-specific host/microbial EV-based signature associated to thrombosis history and marrow fibrosis. These data may contribute to refining PV prognosis and to identifying novel druggable targets.

6.
Oncoimmunology ; 9(1): 1782575, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32923146

RESUMEN

Myelofibrosis (MF) is characterized by chronic inflammation and hyper-activation of the JAK-STAT pathway. Infections are one of the main causes of morbidity/mortality. Therapy with Ruxolitinib (RUX), a JAK1/2 inhibitor, may further increase the infectious risk. Monocytes are critical players in inflammation/immunity through cytokine production and release of bioactive extracellular vesicles. However, the functional behavior of MF monocytes, particularly during RUX therapy, is still unclear. In this study, we found that monocytes from JAK2V617F-mutated MF patients show an altered expression of chemokine (CCR2, CXCR3, CCR5) and cytokine (TNF-α-R, IL10-R, IL1ß-R, IL6-R) receptors. Furthermore, their ability to produce and secrete free and extracellular vesicles-linked cytokines (IL1ß, TNF-α, IL6, IL10) under lipopolysaccharides (LPS) stimulation is severely impaired. Interestingly, monocytes from RUX-treated patients show normal level of chemokine, IL10, IL1ß, and IL6 receptors together with a restored ability to produce intracellular and to secrete extracellular vesicles-linked cytokines after LPS stimulation. Conversely, RUX therapy does not normalize TNF-R1/2 receptors expression and the LPS-driven secretion of free pro/anti-inflammatory cytokines. Accordingly, upon LPS stimulation, in vitro RUX treatment of monocytes from MF patients increases their secretion of extracellular vesicles-linked cytokines but inhibits the secretion of free pro/anti-inflammatory cytokines. In conclusion, we demonstrated that in MF the infection-driven response of circulating monocytes is defective. Importantly, RUX promotes their infection-driven cytokine production suggesting that infections following RUX therapy may not be due to monocyte failure. These findings contribute to better interpreting the immune vulnerability of MF and to envisaging strategies to improve the infection-driven immune response.


Asunto(s)
Mielofibrosis Primaria , Citocinas , Humanos , Lipopolisacáridos , Monocitos , Mielofibrosis Primaria/tratamiento farmacológico , Factor de Necrosis Tumoral alfa
7.
Thromb Haemost ; 120(1): 107-120, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31858520

RESUMEN

Hemolytic uremic syndrome (HUS), the leading cause of acute renal failure in children (< 3 years), is mainly related to Shiga toxins (Stx)-producing Escherichia coli (STEC) infections. STEC are confined to the gut resulting in hemorrhagic colitis, whereas Stx are delivered in blood to target kidney and brain, with unclear mechanisms, triggering HUS in 5 to 15% of infected children. Stx were found on circulating cells, free in sera (soluble Stx) or in blood cell-derived microvesicles (particulate Stx), whereby the relationship between these forms of circulating toxins is unclear. Here, we have examined 2,846 children with bloody diarrhea and found evidence of STEC infection in 5%. Twenty patients were enrolled to study the natural course of STEC infections before the onset of HUS. In patients, Stx were found to be associated to circulating cells and/or free and functionally active in sera. In most children, Stx were bound to neutrophils when high amounts of toxins were found in feces. Time-course analysis showed that Stx increased transiently in patients' sera while the decrease of toxin amount on leukocytes was observed. Notably, patients who recovered (85%) displayed different settings than those who developed HUS (15%). The distinctive feature of the latter group was the presence in blood of particulate Stx2 (Stx2 sedimented at g-forces corresponding to 1 µm microvesicles) the day before diagnosis of HUS, during the release phase of toxins from circulating cells. This observation strongly suggests the involvement of blood cell-derived particulate Stx2 in the transition from hemorrhagic colitis to HUS.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Síndrome Hemolítico-Urémico/metabolismo , Riñón/metabolismo , Neutrófilos/metabolismo , Material Particulado/sangre , Toxina Shiga II/sangre , Escherichia coli Shiga-Toxigénica/fisiología , Adolescente , Línea Celular , Niño , Preescolar , ADN Bacteriano/genética , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Riñón/patología , Masculino , Toxina Shiga II/genética
8.
Cell Microbiol ; 21(5): e13000, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30578712

RESUMEN

Hemolytic uremic syndrome (eHUS) is a severe complication of human infections with Shiga toxins (Stxs)-producing Escherichia coli. A key step in the pathogenesis of eHUS is the interaction of Stxs with blood components before the targeting of renal endothelial cells. Here, we show that a single proteolytic cleavage in the Stx2a A-subunit, resulting into two fragments (A1 and A2) linked by a disulfide bridge (cleaved Stx2a), dictates different binding abilities. Uncleaved Stx2a was confirmed to bind to human neutrophils and to trigger leukocyte/platelet aggregate formation, whereas cleaved Stx2a was ineffective. Conversely, binding of complement factor H was confirmed for cleaved Stx2a and not for uncleaved Stx2a. It is worth noting that uncleaved and cleaved Stx2a showed no differences in cytotoxicity for Vero cells or Raji cells, structural conformation, and contaminating endotoxin. These results have been obtained by comparing two Stx2a batches, purified in different laboratories by using different protocols, termed Stx2a(cl; cleaved toxin, Innsbruck) and Stx2a(uncl; uncleaved toxin, Bologna). Stx2a(uncl) behaved as Stx2a(cl) after mild trypsin treatment. In this light, previous controversial results obtained with purified Stx2a has to be critically re-evaluated; furthermore, characterisation of the structure of circulating Stx2a is mandatory to understand eHUS-pathogenesis and to develop therapeutic approaches.


Asunto(s)
Escherichia coli/química , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Chlorocebus aethiops , Dicroismo Circular , Factor H de Complemento/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorescencia , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Unión Proteica , Conformación Proteica , Toxina Shiga II/genética , Trihexosilceramidas/metabolismo , Tripsina , Células Vero
9.
Toxins (Basel) ; 10(9)2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231570

RESUMEN

Shiga toxin 2a (Stx2a) is the main virulence factor produced by pathogenic Escherichia coli strains (Stx-producing E. coli, STEC) responsible for hemorrhagic colitis and the life-threatening sequela hemolytic uremic syndrome in children. The toxin released in the intestine by STEC targets the globotriaosylceramide receptor (Gb3Cer) present on the endothelial cells of the brain and the kidney after a transient blood phase during which Stx2a interacts with blood components, such as neutrophils, which, conversely, recognize Stx through Toll-like receptor 4 (TLR4). Among non-cellular blood constituents, human amyloid P component (HuSAP) is considered a negative modulating factor that specifically binds Stx2a and impairs its toxic action. Here, we show that the soluble extracellular domain of TLR4 inhibits the binding of Stx2a to neutrophils, assessed by indirect flow cytometric analysis. Moreover, by using human sensitive Gb3Cer-expressing cells (Raji cells) we found that the complex Stx2a/soluble TLR4 escaped from capture by HuSAP allowing the toxin to target and damage human cells, as assayed by measuring translation inhibition, the typical Stx-induced functional impairment. Thus, soluble TLR4 stood out as a positive modulating factor for Stx2a. In the paper, these findings have been discussed in the context of the pathogenesis of hemolytic uremic syndrome.


Asunto(s)
Componente Amiloide P Sérico/metabolismo , Toxina Shiga II/toxicidad , Receptor Toll-Like 4/metabolismo , Línea Celular Tumoral , Humanos , Neutrófilos/metabolismo , Dominios Proteicos
10.
Stem Cell Res Ther ; 8(1): 31, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28173875

RESUMEN

BACKGROUND: The study of lipid metabolism in stem cell physiology has recently raised great interest. The role of lipids goes beyond the mere structural involvement in assembling extra- and intra-cellular compartments. Nevertheless, we are still far from understanding the impact of membrane lipidomics in stemness maintenance and differentiation patterns. In the last years, it has been reported how in vitro cell culturing can modify membrane lipidomics. The aim of the present work was to study the membrane fatty acid profile of mesenchymal stromal cells (MSCs) derived from human fetal membranes (hFM-MSCs) and to correlate this to specific biological properties by using chemically defined tailored lipid supplements (Refeed®). METHODS: Freshly isolated hFM-MSCs were characterized for their membrane fatty acid composition. hFM-MSCs were cultivated in vitro following a classical protocol and their membrane fatty acid profile at different passages was compared to the profile in vivo. A tailored Refeed® lipid supplement was developed with the aim of reducing the differences created by the in vitro cultivation and was tested on cultured hFM-MSCs. Cell morphology, viability, proliferation, angiogenic differentiation, and immunomodulatory properties after in vitro exposure to the tailored Refeed® lipid supplement were investigated. RESULTS: A significant modification of hFM-MSC membrane fatty acid composition occurred during in vitro culture. Using a tailored lipid supplement, the fatty acid composition of cultured cells remained more similar to their in vivo counterparts, being characterized by a higher polyunsaturated and omega-6 fatty acid content. These changes in membrane composition had no effect on cell morphology and viability, but were linked with increased cell proliferation rate, angiogenic differentiation, and immunomodulatory properties. In particular, Refeed®-supplemented hFM-MSCs showed greater ability to express fully functional cell membrane molecules. CONCLUSIONS: Culturing hFM-MSCs alters their fatty acid composition. A tailored lipid supplement is able to improve in vitro hFM-MSC functional properties by recreating a membrane environment more similar to the physiological counterpart. This approach should be considered in cell therapy applications in order to maintain a higher cell quality during in vitro passaging and to influence the outcome of cell-based therapeutic approaches when cells are administered to patients.


Asunto(s)
Antioxidantes/farmacología , Membrana Celular/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Diferenciación Celular , Membrana Celular/química , Proliferación Celular , Suplementos Dietéticos , Membranas Extraembrionarias/citología , Membranas Extraembrionarias/efectos de los fármacos , Membranas Extraembrionarias/metabolismo , Ácidos Grasos/análisis , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/análisis , Ácidos Grasos Monoinsaturados/metabolismo , Ácidos Grasos Insaturados/análisis , Ácidos Grasos Insaturados/metabolismo , Femenino , Humanos , Lípidos de la Membrana/análisis , Lípidos de la Membrana/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Placenta/citología , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Cultivo Primario de Células
11.
J Immunol ; 196(3): 1177-85, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26695372

RESUMEN

Hemolytic uremic syndrome (HUS) is the life-threatenig sequela of intestinal infections by Shiga toxin (Stx)-producing Escherichia coli (STEC) in children. Human neutrophils specifically bind Stx through TLR4, the receptor of LPS. The binding could be considered protective (Stx sequestration) or harmful (toxin delivery to target organs). The amount of Stx on neutrophils is in equilibrium with the amount of Stx present in the gut, and it is also related to renal and neurologic symptoms. The TLR4-mediated interaction of LPS with innate immune cells is hampered by the well-known antibiotic polymyxin B. In this study, we show that the same antibiotic impairs the binding of Stx to neutrophils, also blocking their functional effects (release of CXCL8, formation of neutrophil/platelet aggregates) involved in HUS pathogenesis. Controls for contaminating LPS in Stx-induced neutrophil responses inhibited by polymyxin B were performed. Stx interact with human neutrophils through their A chain, since these leukocytes do not express globotriaosylceramide, the specific receptor for Stx B chains. Consistently, polymyxin B blocked the enzymatic activity of Stx1, Stx2, Stx1 A chain, and the analogous plant protein gelonin, whereas the antibiotic did not show any protective effect on Stx-induced cytotoxicity in globotriaosylceramide-expressing Raji cells. Antibiotic administration is not recommended in human STEC infections during the prodromal intestinal phase, and the toxicity of polymyxin B could further discourage its therapeutic use. However, nontoxic, nonbactericidal polymyxin derivatives have been developed and might be used in animal models of STEC infection to study their efficacy in preventing the onset of HUS during the systemic blood phase of Stx.


Asunto(s)
Antibacterianos/farmacología , Síndrome Hemolítico-Urémico/inmunología , Neutrófilos/efectos de los fármacos , Polimixina B/farmacología , Toxina Shiga/toxicidad , Animales , Citometría de Flujo , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Humanos , Ratones , Neutrófilos/inmunología
12.
Blood Transfus ; 12(4): 458-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24960642

RESUMEN

INTRODUCTION: Assessing blood-donor haemoglobin (Hb) is a worldwide screening requirement against inappropriate donation. The pre-donation Hb (which should be at least 12.5 g/dL in women and 13.5 g/dL in men) is usually determined in capillary blood from a finger prick, using a spectrophotometer which reveals the absorbance of blood haemolysed in a microcuvette. New non-invasive methods of measuring Hb are now available. MATERIALS AND METHODS: In the first semester of 3 consecutive years three different strategies were employed to screen donors for anaemia at the moment of donation. In 2011 all whole-blood donors underwent the finger-prick method using azide-methaemoglobin: the test's negative predictive value (NPV) was determined by comparison with the sub-threshold Hb values ascertained by haemocytometry of test-tube blood drawn at the start of the donation. In 2012 the donor evaluation was based on NBM 200 occlusion spectrophotometry. The same approach was kept in 2013, but a haemocytometry test was added on a pre-donation venous sample drawn from donors who, though fit to donate, had previous critical Hb values in their clinical records. RESULTS: In 2011, the NPV (in 3,856 donors) was 86% for women and 95% for men; in 2012 (3,966 donors), the values were 85% and 95%, respectively, and in 2013 (3,995 donors) they were 91% and 97%, respectively. Fisher's test for contingency tables revealed no statistically significant differences between 2011 and 2012, but the 2013 results were a significant improvement. DISCUSSION: Measuring Hb by finger prick is not wholly satisfactory since, above all in women, the result of this screening may subsequently be belied by the haemocytometry finding of an unacceptable Hb value. Using a non-invasive method does not diminish the selective efficiency. In women, in particular, adding a haemocytometric test on a venous sample significantly improves donor selection and avoids the risk of inappropriate donation or blood-letting.


Asunto(s)
Anemia/sangre , Donantes de Sangre , Selección de Donante/métodos , Hemoglobinas/metabolismo , Femenino , Hemoglobinometría/instrumentación , Hemoglobinometría/métodos , Humanos , Masculino
14.
Adv Biol Regul ; 56: 6-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24819383

RESUMEN

Leukemia initiating cells (LICs) represent a reservoir that is believed to drive relapse and resistance to chemotherapy in blood malignant disorders. T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive neoplastic disorder of immature hematopoietic precursors committed to the T-cell lineage. T-ALL comprises about 15% of pediatric and 25% of adult ALL cases and is prone to early relapse. Although the prognosis of T-ALL has improved especially in children due to the use of new intensified treatment protocols, the outcome of relapsed T-ALL cases is still poor. Putative LICs have been identified also in T-ALL. LICs are mostly quiescent and for this reason highly resistant to chemotherapy. Therefore, they evade treatment and give rise to disease relapse. At present great interest surrounds the development of targeted therapies against signaling networks aberrantly activated in LICs and important for their survival and drug-resistance. Both the Notch1 pathway and the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) network are involved in T-ALL LIC survival and drug-resistance and could be targeted by small molecules. Thus, Notch1 and PI3K/Akt/mTOR inhibitors are currently being developed for clinical use either as single agents or in combination with conventional chemotherapy for T-ALL patient treatment. In this review, we summarize the existing knowledge of the relevance of Notch1 and PI3K/Akt/mTOR signaling in T-ALL LICs and we examine the rationale for targeting these key signal transduction networks by means of selective pharmacological inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transducción de Señal/efectos de los fármacos , Animales , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
15.
PLoS One ; 9(2): e89497, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586826

RESUMEN

BACKGROUND: Neutrophil gelatinase-associated lipocalin (NGAL) is emerging as a mediator of various biological and pathological states. However, the specific biological role of this molecule remains unclear, as it serves as a biomarker for many conditions. The high sensitivity of NGAL as a biomarker coupled with relatively low specificity may hide important biological roles. Data point toward an acute compensatory, protective role for NGAL in response to adverse cellular stresses, including inflammatory and oxidative stress. The aim of this study was to understand whether NGAL modulates the T-cell response through regulation of the human leukocyte antigen G (HLA-G) complex, which is a mediator of tolerance. METHODOLOGY/PRINCIPAL FINDINGS: Peripheral blood mononuclear cells (PBMCs) were obtained from eight healthy donors and isolated by centrifugation on a Ficoll gradient. All donors gave informed consent. PBMCs were treated with four different concentrations of NGAL (40-320 ng/ml) in an iron-loaded or iron-free form. Changes in cell phenotype were analyzed by flow cytometry. NGAL stimulated expression of HLA-G on CD4+ T cells in a dose- and iron-dependent manner. Iron deficiency prevented NGAL-mediated effects, such that HLA-G expression was unaltered. Furthermore, NGAL treatment affected stimulation of regulatory T cells and in vitro expansion of CD4(+) CD25(+) FoxP3(+) cells. An NGAL neutralizing antibody limited HLA-G expression and significantly decreased the percentage of CD4(+) CD25(+) FoxP3(+) cells. CONCLUSIONS/SIGNIFICANCE: We provide in vitro evidence that NGAL is involved in cellular immunity. The potential role of NGAL as an immunomodulatory molecule is based on its ability to induce immune tolerance by upregulating HLA-G expression and expansion of T-regulatory cells in healthy donors. Future studies should further evaluate the role of NGAL in immunology and immunomodulation and its possible relationship to immunosuppressive therapy efficacy, tolerance induction in transplant patients, and other immunological disorders.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Lipocalinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Proteínas de Fase Aguda/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Enterobactina/farmacología , Factores de Transcripción Forkhead/metabolismo , Antígenos HLA-G/metabolismo , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipocalina 2 , Lipocalinas/farmacología , Activación de Linfocitos/efectos de los fármacos , Proteínas Proto-Oncogénicas/farmacología , Subgrupos de Linfocitos T , Linfocitos T Reguladores/efectos de los fármacos
16.
Stem Cell Res Ther ; 5(1): 8, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24429026

RESUMEN

INTRODUCTION: Regenerative medicine challenges researchers to find noncontroversial, safe and abundant stem cell sources. In this context, harvesting from asystolic donors could represent an innovative and unlimited reservoir of different stem cells. In this study, cadaveric vascular tissues were established as an alternative source of human cadaver mesenchymal stromal/stem cells (hC-MSCs). We reported the successful cell isolation from postmortem arterial segments stored in a tissue-banking facility for at least 5 years. METHODS: After thawing, hC-MSCs were isolated with a high efficiency (12×106) and characterized with flow cytometry, immunofluorescence, molecular and ultrastructural approaches. RESULTS: In early passages, hC-MSCs were clonogenic, highly proliferative and expressed mesenchymal (CD44, CD73, CD90, CD105, HLA-G), stemness (Stro-1, Oct-4, Notch-1), pericyte (CD146, PDGFR-ß, NG2) and neuronal (Nestin) markers; hematopoietic and vascular markers were negative. These cells had colony and spheroid-forming abilities, multipotency for their potential to differentiate in multiple mesengenic lineages and immunosuppressive activity to counteract proliferation of phytohemagglutinin-stimulated blood mononuclear cells. CONCLUSIONS: The efficient procurement of stem cells from cadaveric sources, as postmortem vascular tissues, demonstrates that such cells can survive to prolonged ischemic insult, anoxia, freezing and dehydration injuries, thus paving the way for a scientific revolution where cadaver stromal/stem cells could effectively treat patients demanding cell therapies.


Asunto(s)
Arterias/citología , Criopreservación , Células Madre Mesenquimatosas/citología , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Antígenos/genética , Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Arterias/efectos de los fármacos , Cadáver , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Endoglina , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Antígenos HLA-G/genética , Antígenos HLA-G/metabolismo , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Nestina/genética , Nestina/metabolismo , Nitrógeno/farmacología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Investigación con Células Madre , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Recolección de Tejidos y Órganos/métodos
17.
J Tissue Eng Regen Med ; 8(6): 483-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22715158

RESUMEN

Mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP) are currently used alone or in combination for therapeutic applications especially for bone repair. We tested whether MSCs can be isolated from bone marrow (BM) aspirate using a commercially available kit commonly used to obtain PRP from peripheral blood (PB). Results revealed that mononuclear cells and platelets from both PB and BM could be efficiently isolated by obtaining a mononuclear and platelet rich fraction (PB-MPRF and BM-MPRF, respectively). Starting with comparable volumes, the number of platelets increased 1.5-fold in BM-MPRF compared to PB-MPRF. The number of clonogenic cells in BM-MPRF samples was significantly higher than whole BM samples as revealed by CFU-F assay (54.92 ± 8.55 CFU-F/1.5 x 10(5) nucleated cells and 32.50 ± 12.43 CFU-F/1.5 x 10(5) nucleated cells, respectively). Cells isolated from BM-MPRF after in vitro expansion fulfilled the definition of MSCs by phenotypic criteria, and differentiated along osteogenic, adipogenic and chondrogenic lineages following induction. Results showed that the kit isolated MSCs and platelets from BM aspirate. Isolated MSCs were further expanded in a laboratory and BM-MPRF was used clinically following BM withdrawal for rapid intra-operative cell therapy for the treatment of bone defects.


Asunto(s)
Plaquetas/citología , Células de la Médula Ósea/citología , Separación Celular/métodos , Células Madre Mesenquimatosas/citología , Adolescente , Antígenos CD/metabolismo , Biopsia con Aguja , Diferenciación Celular , Niño , Condrogénesis , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Humanos , Osteogénesis , Recuento de Plaquetas , Adulto Joven
18.
J Immunol ; 191(9): 4748-58, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24068665

RESUMEN

Hemolytic uremic syndrome (HUS) caused by intestinal Shiga toxin-producing Escherichia coli infections is a worldwide health problem, as dramatically exemplified by the German outbreak occurred in summer 2011 and by a constant burden of cases in children. Shiga toxins (Stx) play a pivotal role in HUS by triggering endothelial damage in kidney and brain through globotriaosylceramide (Gb3Cer) receptor targeting. Moreover, Stx interact with human neutrophils, as experimentally demonstrated in vitro and as observed in patients with HUS. A neutrophil-protective role on endothelial damage (sequestration of circulating toxins) and a causative role in toxin delivery from the gut to the kidney (piggyback transport) have been suggested in different studies. However, the receptor that recognizes Stx in human neutrophils, which do not express Gb3Cer, has not been identified. In this study, by competition and functional experiments with appropriate agonists and antagonists (LPS, anti-TLR4 Abs, respectively), we have identified TLR4 as the receptor that specifically recognizes Stx1 and Stx2 in human neutrophils. Accordingly, these treatments displaced both toxin variants from neutrophils and, upon challenge with Stx1 or Stx2, neutrophils displayed the same pattern of cytokine expression as in response to LPS (assessed by quantitative RT-PCR, ELISA, or multiplexed Luminex-based immunoassays). Moreover, data were supported by adequate controls excluding any potential interference of contaminating LPS in Stx-binding and activation of neutrophils. The identification of the Stx-receptor on neutrophils provides additional elements to foster the understanding of the pathophysiology of HUS and could have an important effect on the development of therapeutic strategies.


Asunto(s)
Neutrófilos/metabolismo , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Receptor Toll-Like 4/inmunología , Anticuerpos Monoclonales , Citocinas/metabolismo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Lipopolisacáridos , Neutrófilos/inmunología , Trihexosilceramidas/metabolismo
19.
Toxins (Basel) ; 5(2): 431-44, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23430607

RESUMEN

Shiga toxin 1 (Stx1), produced by pathogenic Escherichia coli, targets a restricted subset of human cells, which possess the receptor globotriaosylceramide (Gb3Cer/CD77), causing hemolytic uremic syndrome. In spite of the high toxicity, Stx1 has been proposed in the treatment of Gb3Cer/CD77-expressing lymphoma. Here, we demonstrate in a Burkitt lymphoma cell model expressing this receptor, namely Raji cells, that Stx1, at quasi-non-toxic concentrations (0.05-0.1 pM), inhibits the repair of mafosfamide-induced DNA alkylating lesions, synergistically potentiating the cytotoxic activity of the anticancer drug. Conversely, human promyelocytic leukemia cells HL-60, which do not express Gb3Cer/CD77, were spared by the toxin as previously demonstrated for CD34+ human progenitor cells, and hence, in this cancer model, no additive nor synergistic effects were observed with the combined Stx1/mafosfamide treatment. Our findings suggest that Stx1 could be used to improve the mafosfamide-mediated purging of Gb3Cer/CD77+ tumor cells before autologous bone marrow transplantation.


Asunto(s)
Antineoplásicos/administración & dosificación , Ciclofosfamida/análogos & derivados , Inhibidores de la Síntesis de la Proteína/administración & dosificación , Toxina Shiga I/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclofosfamida/administración & dosificación , Reparación del ADN/efectos de los fármacos , Sinergismo Farmacológico , Células HL-60 , Células Endoteliales de la Vena Umbilical Humana , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...