Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 13: 220, 2015 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-26160352

RESUMEN

BACKGROUND: The intestinal stem cells (ISC) modulation and the role of circulating hematopoietic stem cells (HSC) in coeliac disease (CD) are poorly understood. Our aim was to investigate the longitudinal modifications in peripheral blood HSC traffic and putative ISC density induced by gluten-free diet (GFD) in CD. METHODS: Thirty-one CD patients and 7 controls were enrolled. Circulating CD133(+) and CD34(+) HSC were measured by flow cytometry, at enrolment and after 7 days and 1, 3, 6, 12, and 24 months of GFD. Endoscopy was performed at diagnosis and repeated at 6, 12, and 24 months following GFD. We used the Marsh-Oberhuber score to evaluate the histological severity of duodenal damage; immunohistochemistry was employed to measure the intraepithelial lymphoid infiltrate (IEL, CD3(+) lymphoid cells) and the putative ISC compartment (CD133(+) and Lgr5(+) epithelial cells). RESULTS: At enrolment, circulating HSCs were significantly increased in CD patients and they further augmented during the first week of GFD, but progressively decreased afterwards. CD patients presented with villous atrophy, abundant IEL and rare ISC residing at the crypt base. Upon GFD, IEL progressively decreased, while ISC density increased, peaking at 12 months. After 24 months of GFD, all patients were asymptomatic and their duodenal mucosa was macroscopically and histologically normal. CONCLUSIONS: In active CD patients, the ISC niche is depleted and there is an increased traffic of circulating HSC versus non-coeliac subjects. GFD induces a precocious mobilization of circulating HSC, which is followed by the expansion of the local ISC compartment, leading to mucosal healing and clinical remission.


Asunto(s)
Enfermedad Celíaca/sangre , Enfermedad Celíaca/patología , Movimiento Celular , Células Madre Hematopoyéticas/patología , Intestinos/patología , Antígeno AC133 , Adulto , Anciano , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Estudios de Casos y Controles , Recuento de Células , Dieta Sin Gluten , Femenino , Estudios de Seguimiento , Glicoproteínas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Péptidos/metabolismo , Adulto Joven
2.
Transfusion ; 55(12): 2864-73, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26354088

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) isolated from human umbilical cord tissue (UCT) can be considered the perfect candidates for cell-based therapies and regenerative medicine. UCT-derived MSCs can be cryogenically stored in cell banks and expanded as needed for therapeutic uses. STUDY DESIGN AND METHODS: We developed a new method for UCT-MSC isolation, cryopreservation, and expansion, following all criteria required by a stem cell bank. UCT-MSCs were isolated either by manual dissociation (MM) or by a semiautomatic dissociation system (SAM). In both protocols UCTs were treated enzymatically using Type IV collagenase good manufacturing practices (GMP) graded and hyaluronidase (medicinal product). Isolated UCT-MSCs were cryopreserved and analyzed after thawing for phenotype; for proliferation rate; and for their osteogenic, adipogenic, and chondrogenic differentiation capabilities. RESULTS: We found that SAM reduced the time of tissue enzyme exposure and enabled us to obtain a homogeneous single-cell suspension deprived of tissue fragments. The isolated cells in both groups showed high expression of MSC markers CD105, CD73, and CD90 and similar differentiation capabilities, phenotype, and proliferation potential. Moreover, the final yield of MSCs was comparable between the two techniques. CONCLUSION: In this study, we have established a reliable and standardized protocol to isolate UCT-MSCs from UCT for cell banking purposes. Processing the whole umbilical tissue with GMP-graded enzymes using a semiautomatic dissociator allowed us to obtain a single-cell suspension product with a known number of isolated cells that can be cryopreserved right after isolation and thawed as needed for expansion and clinical use.


Asunto(s)
Protocolos Clínicos/normas , Bancos de Tejidos/normas , Cordón Umbilical/citología , Separación Celular/métodos , Criopreservación/métodos , Humanos , Células Madre Mesenquimatosas/citología
3.
J Transl Med ; 12: 28, 2014 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-24467837

RESUMEN

BACKGROUND: Supplements to support clinical-grade cultures of mesenchymal stem cells (MSC) are required to promote growth and expansion of these cells. Platelet lysate (PL) is a human blood component which may replace animal serum in MSC cultures being rich in various growth factors. Here, we describe a plasma poor pathogen-free platelet lysate obtained by pooling 12 platelet (PLT) units, to produce a standardized and safe supplement for clinical-grade expansion of MSC. METHODS: PL lots were obtained by combining 2 6-unit PLT pools in additive solution (AS) following a transfusional-based procedure including pathogen inactivation (PI) by Intercept technology and 3 cycles of freezing/thawing, followed by membrane removal. Three PI-PL and 3 control PL lots were produced to compare their ability to sustain bone marrow derived MSC selection and expansion. Moreover, two further PL, subjected to PI or not, were also produced starting from the same initial PLT pools to evaluate the impact of PI on growth factor concentration and capacity to sustain cell growth. Additional PI-PL lots were used for comparison with fetal bovine serum (FBS) on MSC expansion. Immunoregulatory properties of PI-PL-generated MSC were documented in vitro by mixed lymphocyte culture (MLC) and peripheral blood mononuclear cells (PBMC) mitogen induced proliferation. RESULTS: PI-PL and PL control lots had similar concentrations of 4 well-described growth factors endowed with MSC stimulating ability. Initial growth and MSC expansion by PI-PL and PL controls were comparable either using different MSC populations or in head to head experiments. Moreover, PI-PL and PL control sustained similar MSC growth of frozen/thawed MSC. Multilineage differentiation of PI-derived and PI-PL-derived MSC were maintained in any MSC cultures as well as their immunoregulatory properties. Finally, no direct impact of PI on growth factor concentration and MSC growth support was observed, whereas the capacity of FBS to sustain MSC expansion in basic medium was irrelevant as compared to PL and PI-PL. CONCLUSION: The replacement of animal additives with human supplements is a basic issue in MSC ex vivo production. PI-PL represents a standardized, plasma-poor, human preparation which appears as a safe and good candidate to stimulate MSC growth in clinical-scale cultures.


Asunto(s)
Plaquetas/metabolismo , Extractos Celulares/farmacología , Células Madre Mesenquimatosas/citología , Viabilidad Microbiana , Plasma/metabolismo , Animales , Antígenos/metabolismo , Plaquetas/efectos de los fármacos , Bovinos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Criopreservación , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Terapia de Inmunosupresión , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Viabilidad Microbiana/efectos de los fármacos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
4.
Molecules ; 18(9): 10132-45, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973990

RESUMEN

Indoleamine 2,3-dioxygenase 1 (IDO1) metabolizes L-tryptophan to kynurenines (KYN), inducing T-cell suppression either directly or by altering antigen-presenting-cell function. Cyclooxygenase (COX)-2, the rate-limiting enzyme in the synthesis of prostaglandins, is over-expressed by several tumours. We aimed at determining whether COX-2 inhibitors down-regulate the IFN-g-induced expression of IDO1 in acute myeloid leukaemia (AML) cells. IFN-γ at 100 ng/mL up-regulated COX-2 and IDO1 in HL-60 AML cells, both at mRNA and protein level. The increased COX-2 and IDO1 expression correlated with heightened production of prostaglandin (PG)E2 and kynurenines, respectively. Nimesulide, a preferential COX-2 inhibitor, down-regulated IDO1 mRNA/protein and attenuated kynurenine synthesis, suggesting that overall IDO inhibition resulted both from reduced IDO1 gene transcription and from inhibited IDO1 catalytic activity. From a functional standpoint, IFN-g-challenged HL-60 cells promoted the in vitro conversion of allogeneic CD4⁺CD25⁻ T cells into bona fide CD4⁺CD25⁺FoxP3⁺ regulatory T cells, an effect that was significantly reduced by treatment of IFN-γ-activated HL-60 cells with nimesulide. Overall, these data point to COX-2 inhibition as a potential strategy to be pursued with the aim at circumventing leukaemia-induced, IDO-mediated immune dysfunction.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Sulfonamidas/farmacología , Técnicas de Cocultivo , Ciclooxigenasa 2/genética , Dinoprostona/biosíntesis , Dinoprostona/metabolismo , Inducción Enzimática/efectos de los fármacos , Células HL-60 , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Interferón gamma/fisiología , Quinurenina/biosíntesis , Leucemia Mieloide Aguda , Receptores de Interferón/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Receptor de Interferón gamma
5.
J Transl Med ; 10: 247, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23232072

RESUMEN

BACKGROUND: Multiple myeloma (MM) is a plasma cell malignancy with a multifaceted immune dysfunction. Indoleamine 2,3-dioxygenase 1 (IDO1) degrades tryptophan into kynurenine (KYN), which inhibits effector T cells and promote regulatory T-cell (Treg) differentiation. It is presently unknown whether MM cells express IDO1 and whether IDO1 activity correlates with immune system impairment. METHODS: We investigated IDO1 expression in 25 consecutive patients with symptomatic MM and in 7 patients with either monoclonal gammopathy of unknown significance (MGUS; n=3) or smoldering MM (SMM; n=4). IDO1-driven tryptophan breakdown was correlated with the release of hepatocyte growth factor (HGF) and with the frequency of Treg cells and NY-ESO-1-specific CD8(+) T cells. RESULTS: KYN was increased in 75% of patients with symptomatic MM and correlated with the expansion of CD4(+)CD25(+)FoxP3(+) Treg cells and the contraction of NY-ESO-1-specific CD8(+) T cells. In vitro, primary MM cells promoted the differentiation of allogeneic CD4(+) T cells into bona fide CD4(+)CD25(hi)FoxP3(hi) Treg cells and suppressed IFN-γ/IL-2 secretion, while preserving IL-4 and IL-10 production. Both Treg expansion and inhibition of Th1 differentiation by MM cells were reverted, at least in part, by D,L-1-methyl-tryptophan, a chemical inhibitor of IDO. Notably, HGF levels were higher within the BM microenvironment of patients with IDO(+) myeloma disease compared with patients having IDO(-) MM. Mechanistically, the antagonism of MET receptor for HGF with SU11274, a MET inhibitor, prevented HGF-induced AKT phosphorylation in MM cells and translated into reduced IDO protein levels and functional activity. CONCLUSIONS: These data suggest that IDO1 expression may contribute to immune suppression in patients with MM and possibly other HGF-producing cancers.


Asunto(s)
Sistema Inmunológico/anomalías , Sistema Inmunológico/enzimología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Mieloma Múltiple/enzimología , Mieloma Múltiple/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Proliferación Celular , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Interleucina-10/metabolismo , Proteínas de la Membrana/metabolismo , Células Plasmáticas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/metabolismo , Carga Tumoral/inmunología
6.
Cytotherapy ; 14(7): 841-50, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22563888

RESUMEN

BACKGROUND AIMS: We have recently shown that thymoglobulin (TG) efficiently expands cytokine-induced killer (CIK) cells in combination with interferon (IFN)-γ and interleukin (IL)-2 (ITG2 protocol). It is presently unknown whether the infusion of autologous immune effector cells generated by TG, IFN-γ and IL-2 is feasible and safe. METHODS: Five patients with advanced and/or refractory solid tumors were enrolled in the present phase I/II study. Peripheral blood mononuclear cells (PBMC) collected by leukapheresis were stimulated under good manufacturing practice (GMP)-conditions with IFN-γ, followed by TG and IL-2. After 2-3 weeks in culture, a median of 4.65 × 10(6) immune effector cells per kilogram of recipient's body weight was obtained and infused intravenously. The median time from enrollment into the study to infusion of the expanded CIK cells was 30 days. RESULTS: ITG2 efficiently expanded immune effector cells that comprised both conventional natural killer (NK) cells and CD3(+) CD16(+) CD56(+) CIK cells. One patient with advanced melanoma died because of disease progression before the infusion of CIK cells. The target dose of at least 2.5 × 10(6) CIK cells/kg of recipient's body weight was reached in four out of five evaluable patients. CIK cells were administered intravenously without any measurable toxicity. In vitro, CIK cells exerted lytic activity against cervical cancer cells. The median survival was 4.5 months (range 1-13) from the first infusion of CIK cells. CONCLUSIONS: This study has highlighted the feasibility and safety of the administration of CIK cells generated with the ITG2 protocol. Whether CIK cells can help control disease burden in patients with advanced malignancies will be determined in future clinical trials.


Asunto(s)
Células Asesinas Inducidas por Citocinas , Inmunoterapia Adoptiva , Leucocitos Mononucleares , Melanoma , Neoplasias del Cuello Uterino , Adulto , Suero Antilinfocítico/farmacología , Células Cultivadas , Células Asesinas Inducidas por Citocinas/citología , Células Asesinas Inducidas por Citocinas/efectos de los fármacos , Células Asesinas Inducidas por Citocinas/trasplante , Femenino , Humanos , Interferón gamma/farmacología , Interleucina-2/farmacología , Leucaféresis , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/trasplante , Melanoma/sangre , Melanoma/terapia , Persona de Mediana Edad , Neoplasias del Cuello Uterino/sangre , Neoplasias del Cuello Uterino/terapia
7.
J Transl Med ; 8: 114, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-21062439

RESUMEN

BACKGROUND: Pegylated granulocyte colony-stimulating factor (G-CSF; pegfilgrastim) is a longer-acting form of G-CSF, whose effects on dendritic cell (DC) and regulatory T cell (Treg) mobilization, and on the in vivo and ex vivo release of immune modulating cytokines remain unexplored. METHODS: Twelve patients with gynecological cancers received carboplatin/paclitaxel chemotherapy and single-dose pegfilgrastim as prophylaxis of febrile neutropenia. Peripheral blood was collected prior to pegfilgrastim administration (day 0) and on days +7, +11 and +21, to quantify immunoregulatory cytokines and to assess type 1 DC (DC1), type 2 DC (DC2) and Treg cell mobilization. In vitro-differentiated, monocyte-derived DC were used to investigate endocytic activity, expression of DC maturation antigens and ability to activate allogeneic T-cell proliferation. RESULTS: Pegfilgrastim increased the frequency of circulating DC1 and DC2 precursors. In contrast, CD4+FoxP3+ bona fide Treg cells were unchanged compared with baseline. Serum levels of hepatocyte growth factor and interleukin (IL)-12p40, but not transforming growth factor-ß1 or immune suppressive kynurenines, significantly increased after pegfilgrastim administration. Interestingly, pegfilgrastim fostered in vitro monocytic secretion of IL-12p40 and IL-12p70 when compared with unconjugated G-CSF. Finally, DC populations differentiated in vitro after clinical provision of pegfilgrastim were phenotypically mature, possessed low endocytic activity, and incited a robust T-cell proliferative response. CONCLUSIONS: Pegfilgrastim induced significant changes in immune cell number and function. The enhancement of monocytic IL-12 secretion portends favorable implications for pegfilgrastim administration to patients with cancer, a clinical context where the induction of immune deviation would be highly undesirable.


Asunto(s)
Neoplasias de los Genitales Femeninos/patología , Factor Estimulante de Colonias de Granulocitos/farmacología , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/administración & dosificación , Femenino , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Neoplasias de los Genitales Femeninos/inmunología , Humanos , Persona de Mediana Edad , Paclitaxel/administración & dosificación
8.
J Transl Med ; 8: 129, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21138560

RESUMEN

BACKGROUND: Cytokine-induced killer (CIK) cells are typically differentiated in vitro with interferon (IFN)-γ and αCD3 monoclonal antibodies (mAb), followed by the repeated provision of interleukin (IL)-2. It is presently unknown whether thymoglobulin (TG), a preparation of polyclonal rabbit γ immunoglobulins directed against human thymocytes, can improve the generation efficiency of CIK cells compared with αCD3 mAb in a clinical-grade culture protocol. METHODS: Peripheral blood mononuclear cells (PBMC) from 10 healthy donors and 4 patients with solid cancer were primed with IFN-γ on day 0 and low (50 ng/ml), intermediate (250 ng/ml) and high (500 ng/ml) concentrations of either αCD3 mAb or TG on day 1, and were fed with IL-2 every 3 days for 21 days. Aliquots of cells were harvested weekly to monitor the expression of representative members of the killer-like immunoglobulin receptor (KIR), NK inhibitory receptor, NK activating receptor and NK triggering receptor families. We also quantified the frequency of bona fide regulatory T cells (Treg), a T-cell subset implicated in the down-regulation of anti-tumor immunity, and tested the in vitro cytotoxic activity of CIK cells against NK-sensitive, chronic myeloid leukaemia K562 cells. RESULTS: CIK cells expanded more vigorously in cultures supplemented with intermediate and high concentrations of TG compared with 50 ng/ml αCD3 mAb. TG-driven CIK cells expressed a constellation of NK activating/inhibitory receptors, such as CD158a and CD158b, NKp46, NKG2D and NKG2A/CD94, released high quantities of IL-12p40 and efficiently lysed K562 target cells. Of interest, the frequency of Treg cells was lower at any time-point compared with PBMC cultures nurtured with αCD3 mAb. Cancer patient-derived CIK cells were also expanded after priming with TG, but they expressed lower levels of the NKp46 triggering receptor and NKG2D activating receptor, thus manifesting a reduced ability to lyse K562 cells. CONCLUSIONS: TG fosters the generation of functional CIK cells with no concomitant expansion of tumor-suppressive Treg cells. The culture conditions described herein should be applicable to cancer-bearing individuals, although the differentiation of fully functional CIK cells may be hindered in patients with advanced malignancies.


Asunto(s)
Suero Antilinfocítico/farmacología , Técnicas de Cultivo de Célula/métodos , Células Asesinas Inducidas por Citocinas/citología , Células Asesinas Inducidas por Citocinas/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-2/farmacología , Adulto , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Persona de Mediana Edad , Neoplasias/inmunología , Fenotipo , Conejos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Donantes de Tejidos , Adulto Joven
9.
Haematologica ; 95(8): 1253-60, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20305142

RESUMEN

BACKGROUND: The human hemoglobin switch (HbF-->HbA) takes place in the peri/post-natal period. In adult life, however, the residual HbF (<1%) may be partially reactivated by chemical inducers and/or cytokines such as the kit ligand (KL). MicroRNAs (miRs) play a pivotal role in normal hematopoiesis: downmodulation of miR-221/222 stimulates human erythropoietic proliferation through upmodulation of the kit receptor. DESIGN AND METHODS: We have explored the possible role of kit/KL in perinatal Hb switching by evaluating: i) the expression levels of both kit and kit ligand on CD34(+) cells and in plasma isolated from pre-, mid- and full-term cord blood samples; ii) the reactivation of HbF synthesis in KL-treated unilineage erythroid cell cultures; iii) the functional role of miR-221/222 in HbF production. RESULTS: In perinatal life, kit expression showed a gradual decline directly correlated to the decrease of HbF (from 80-90% to <30%). Moreover, in full-term cord blood erythroid cultures, kit ligand induced a marked increase of HbF (up to 80%) specifically abrogated by addition of the kit inhibitor imatinib, thus reversing the Hb switch. MiR-221/222 expression exhibited rising levels during peri/post-natal development. In functional studies, overexpression of these miRs in cord blood progenitors caused a remarkable decrease in kit expression, erythroblast proliferation and HbF content, whereas their suppression induced opposite effects. CONCLUSIONS: Our studies indicate that human perinatal Hb switching is under control of the kit receptor/miR 221-222 complex. We do not exclude, however, that other mechanisms (i.e. glucocorticoids and the HbF inhibitor BCL11A) may also contribute to the peri/post-natal Hb switch.


Asunto(s)
Hemoglobina Fetal/metabolismo , Hemoglobina A/metabolismo , MicroARNs/fisiología , Factor de Células Madre/fisiología , Adulto , Antígenos CD34/sangre , Benzamidas , Ciclo Celular , Células Cultivadas , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Sangre Fetal/citología , Sangre Fetal/metabolismo , Citometría de Flujo , Expresión Génica , Humanos , Mesilato de Imatinib , Recién Nacido , MicroARNs/genética , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-kit/sangre , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/sangre , Factor de Células Madre/genética , Factores de Tiempo
10.
BMC Immunol ; 10: 46, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19712464

RESUMEN

BACKGROUND: Umbilical cord blood (UCB) is enriched with transplantable CD34+ cells. In addition to CD34-expressing haematopoietic stem cells (HSC), human UCB contains a rare population of CD34-lineage- cells endowed with the ability to differentiate along the T/NK pathway in response to interleukin (IL)-15 and a stromal cell support. IL-21 is a crucial regulator of NK cell function, whose influence on IL-15-induced differentiation of CD34-lineage- cells has not been investigated previously. The present study was designed and conducted to address whether IL-21 might replace the stromal cell requirements and foster the IL-15-induced NK differentiation of human UCB CD34-lineage- cells. RESULTS: CD34-lineage- cells were maintained in liquid culture with Flt3-L and SCF, with the addition of IL-15 and IL-21, either alone or in combination. Cultures were established in the absence of feeder cells or serum supplementation. Cytokine-treated cells were used to evaluate cell surface phenotype, expression of molecular determinants of lymphoid/NK cell differentiation, secretion of IFN-gamma, GM-CSF, TNF-alpha and CCL3/MIP-1alpha, and cytolytic activity against NK-sensitive tumour cell targets. CD34-lineage- cells proliferated vigorously in response to IL-15 and IL-21 but not to IL-21 alone, and up-regulated phosphorylated Stat1 and Stat3 proteins. CD34-lineage- cells expanded by IL-21 in combination with IL-15 acquired lymphoid morphology and killer-cell immunoglobulin-like receptor (KIR)-CD56+CD16-/+ phenotype, consistent with pseudo-mature NK cells. IL-21/IL-15-differentiated cells expressed high levels of mRNA for Bcl-2, GATA-3 and Id2, a master switch required for NK-cell development, and harboured un-rearranged TCRgamma genes. From a functional standpoint, IL-21/IL-15-treated cells secreted copious amounts of IFN-gamma, GM-CSF and CCL3/MIP-1alpha, and expressed cell surface CD107a upon contact with NK-sensitive tumour targets, a measure of exocytosis of NK secretory granules. CONCLUSION: This study underpins a novel role for IL-21 in the differentiation of pseudo-mature lytic NK cells in a synergistic context with IL-15, and identifies a potential strategy to expand functional NK cells for immunotherapy.


Asunto(s)
Linaje de la Célula/inmunología , Sangre Fetal/inmunología , Interleucina-15/farmacología , Interleucinas/inmunología , Células Asesinas Naturales/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/fisiología , Diferenciación Celular , Quimiocina CCL3/biosíntesis , Quimiocina CCL3/inmunología , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Factor de Transcripción GATA3/inmunología , Factor de Transcripción GATA3/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Proteína 2 Inhibidora de la Diferenciación/inmunología , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucinas/farmacología , Células Asesinas Naturales/trasplante , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción STAT1/agonistas , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/agonistas , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo
11.
Exp Hematol ; 34(1): 54-65, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413391

RESUMEN

OBJECTIVE: To evaluate the effects of granulocyte colony-stimulating factor (G-CSF) on the in vitro sensitivity of acute myeloid leukemia (AML) cell lines and primary AML blast cells to gemtuzumab ozogamicin (GO). MATERIALS AND METHODS: AML cell lines and primary blasts from 10 patients with AML were first incubated for 72 hours in the presence of G-CSF (5 or 100 ng/mL) and then exposed to increasing concentrations of GO (1-1,000 ng/mL) for an additional 72 hours. RESULTS: Pretreatment with G-CSF translated into significant enhancement of GO-induced cytotoxicity in the GO-sensitive HL-60 and NB-4 cells. Conversely, the response of GO-insensitive KG-1a, TF-1, and K562 cells was unaffected by in vitro priming with G-CSF. In vitro exposure to G-CSF augmented GO-induced apoptosis in 7 of 10 primary AML samples and rendered blast cells from three refractory patients sensitive to killing effect of GO. The G-CSF-induced increase of the cytocidal activity of GO was independent of effects on the cell cycle and on the expression levels of CD33 antigen. Of potential interest, G-CSF induced dose-dependent inhibition of P-glycoprotein (P-gp/ABCB1) function in the GO-sensitive HL-60 and NB-4 cells and in blasts from three patients with AML that we tested. CONCLUSION: Collectively, our findings point to G-CSF as a potential sensitizing agent that can be exploited therapeutically to improve the clinical efficacy of GO.


Asunto(s)
Aminoglicósidos/farmacología , Anticuerpos Monoclonales/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Enfermedad Aguda , Anciano , Anticuerpos Monoclonales Humanizados , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/efectos de los fármacos , Antígenos de Diferenciación Mielomonocítica/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Gemtuzumab , Células HL-60 , Humanos , Técnicas In Vitro , Leucemia Mieloide/diagnóstico , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Factores de Tiempo , Células Tumorales Cultivadas
12.
Int J Cardiol ; 249: 349-356, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-28967436

RESUMEN

Peripheral arterial disease (PAD), is a major health problem. Many studies have been focused on the possibilities of treatment offered by vascular regeneration. Human adipose-derived stem cells (HASCs), multipotent CD34+ stem cells found in the stromal-vascular fraction of adipose tissues, which are capable to differentiate into multiple mesenchymal cell types. The High mobility group box 1 protein (HMGB1) is a nuclear protein involved in angiogenesis. The aim of the study was to define the role of HMGB1 in cell therapy with HASCs, in an animal model of PAD. We induced unilateral ischemia in mice and we treated them with HASCs, with the specific HMGB1-inihibitor BoxA, with HMGB1 protein, and with the specific VEGF inhibitor sFlt1, alternately or concurrently. We measured the blood flow recovery in all mice. Immunohistochemical and ELISA analyses was performed to evaluate the number of vessels and the VEGF tissue content. None auto-amputation occurred and there have been no rejection reactions to the administration of HASCs. Animals co-treated with HASCs and HMGB1 protein had an improved blood flow recovery, compared to HASCs-treated mice. The post-ischemic angiogenesis was reduced when the HMGB1 pathway was blocked or when the VEGF activity was inhibited, in mice co-treated with HASCs and HMGB1. In conclusion, the HASCs treatment can be used in a mouse model of PAD to induce post-ischemic angiogenesis, modulating angiogenesis by HMGB1. This effect is mediated by VEGF activity. Although further data are needed, these findings shed light on possible new cell treatments for patients with PAD.


Asunto(s)
Tejido Adiposo/fisiología , Tejido Adiposo/trasplante , Proteína HMGB1/fisiología , Neovascularización Fisiológica/fisiología , Trasplante de Células Madre/métodos , Adipocitos/fisiología , Adipocitos/trasplante , Animales , Células Cultivadas , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Humanos , Isquemia/patología , Isquemia/fisiopatología , Isquemia/terapia , Ratones , Ratones Endogámicos C57BL , Enfermedad Arterial Periférica/patología , Enfermedad Arterial Periférica/fisiopatología , Enfermedad Arterial Periférica/terapia , Flujo Sanguíneo Regional/fisiología
13.
Circ Res ; 93(5): e51-62, 2003 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-12919944

RESUMEN

Human umbilical cord blood (UCB) contains high numbers of endothelial progenitors cells (EPCs) characterized by coexpression of CD34 and CD133 markers. Prior studies have shown that CD34+/CD133+ EPCs from the cord or peripheral blood (PB) can give rise to endothelial cells and induce angiogenesis in ischemic tissues. In the present study, it is shown that freshly isolated human cord blood CD34+ cells injected into ischemic adductor muscles gave rise to endothelial and, unexpectedly, to skeletal muscle cells in mice. In fact, the treated limbs exhibited enhanced arteriole length density and regenerating muscle fiber density. Under similar experimental conditions, CD34- cells did not enhance the formation of new arterioles and regenerating muscle fibers. In nonischemic limbs CD34+ cells increased arteriole length density but did not promote formation of new muscle fibers. Endothelial and myogenic differentiation ability was maintained in CD34+ cells after ex vivo expansion. Myogenic conversion of human cord blood CD34+ cells was also observed in vitro by coculture onto mouse myoblasts. These results show that human cord blood CD34+ cells differentiate into endothelial and skeletal muscle cells, thus providing an indication of human EPCs plasticity. The full text of this article is available online at http://www.circresaha.org.


Asunto(s)
Diferenciación Celular , Endotelio Vascular/citología , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Miembro Posterior/irrigación sanguínea , Músculo Esquelético/citología , Animales , Antígenos CD34/análisis , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Sangre Fetal/química , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/química , Humanos , Inmunohistoquímica , Recién Nacido , Isquemia/terapia , Ratones , Músculo Esquelético/fisiología , Regeneración , Trasplante Heterólogo
14.
Int J Cardiol ; 111(2): 202-8, 2006 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-16051386

RESUMEN

BACKGROUND: Several reports showed an increase of CD34(+) stem/progenitor cell count early after an acute myocardial infarction (AMI), suggesting a contribution of bone marrow cells in myocardial regeneration after the acute event. Nevertheless, at present plasma mediators of CD34(+) cell mobilization from bone marrow to peripheral blood in patients with AMI are poorly understood. Aim of our study was to establish the impact of different well-known mobilizing cytokines on spontaneous stem cell mobilization in patients with different ischemic heart syndromes, such as the AMI and the chronic stable angina (CSA), compared to healthy controls. METHODS: In 16 patients with AMI, 18 with CSA and 22 healthy blood donors the concentration of CD34(+) cells, and mobilizing cyokines (G-CSF, SCF, VEGF, SDF1-alpha) were assessed. RESULTS: The peak number of circulating CD34(+) cells in AMI patients (8.58+/-2.08 cells/microl) was higher than that observed in patients with CSA (3.41+/-0.56 cells/microl, p=0.0061) or in healthy controls (2.18+/-0.35 cells/microl, p<0.001). However endogenous G-CSF was significantly higher in the serum of patients with AMI compared to CSA patients and to controls and in CSA patients compared to controls. Interestingly, as regards VEGF, while this cytokine was increased in AMI with respect to control and CSA group, the latter showed a significantly lower concentration with respect to controls. Finally SDF-1 alpha was higher in AMI patients with respect to controls. CD34(+) cells were significantly correlated to G-CSF (directly) and to SCF (inversely) in patients with AMI. CONCLUSION: In the present study, we have demonstrated for the first time that the spontaneous mobilization of CD34(+) cells into the peripheral blood of patients with AMI is significantly correlated to endogenous G-CSF. Considering recent data suggesting a potential favourable effect of circulating CD34(+) cells on left ventricular function, the present evidence of a correlation between endogenous G-CSF and CD34(+) cell levels supports the pharmacological administration of G-CSF as a non-invasive option for regeneration of myocardial tissue after AMI.


Asunto(s)
Antígenos CD34/sangre , Factor Estimulante de Colonias de Granulocitos/sangre , Movilización de Célula Madre Hematopoyética , Infarto del Miocardio/sangre , Anciano , Antígenos CD/sangre , Quimiocina CXCL12 , Quimiocinas CXC/sangre , Citocinas/sangre , Femenino , Estudios de Seguimiento , Corazón/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Valores de Referencia , Regeneración
15.
FASEB J ; 18(14): 1737-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15345695

RESUMEN

Hematopoietic progenitor cell transplantation can contribute to revascularization of ischemic tissues. Yet, the optimal cell population to be transplanted has yet to be determined. We have compared the therapeutic potential of two subsets of human cord blood CD34+ progenitors, either expressing the VEGF-A receptor 2 (KDR) or not. In serum-free starvation culture, CD34+KDR+ cells reportedly showed greater resistance to apoptosis and ability to release VEGF-A, as compared with CD34+KDR- cells. When injected into the hind muscles in immunodeficient SCIDbg mice subjected to unilateral ischemia, a low number (10(3)) of CD34+KDR+ cells improved limb salvage and hemodynamic recovery better than a larger dosage (10(4)) of CD34+KDR- cells. The neovascularization induced by KDR+ cells was significantly superior to that promoted by KDR- cells. Similarly, endothelial cell apoptosis and interstitial fibrosis were significantly attenuated by KDR+ cells, which differentiated into mature human endothelial cells and also apparently skeletal muscle cells. This study demonstrates that a low number of CD34+KDR+ cells favors reparative neovascularization and possibly myogenesis in limb ischemia, suggesting the potential use of this cell population in regenerative medicine.


Asunto(s)
Antígenos CD34/análisis , Isquemia/terapia , Músculo Esquelético/fisiología , Neovascularización Fisiológica , Regeneración , Trasplante de Células Madre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Animales , Apoptosis , Extremidades/irrigación sanguínea , Sangre Fetal/citología , Fibrosis , Hemodinámica , Humanos , Isquemia/patología , Isquemia/fisiopatología , Ratones , Fibras Musculares Esqueléticas/fisiología , Células Madre/química
16.
Leuk Lymphoma ; 43(3): 493-501, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12002751

RESUMEN

Hematopoietic stem cells (HSCs) reside mostly in the bone marrow and are defined by their ability to self-renew and to give rise by proliferation and differentiation to all blood lineages. Despite this strict definition HSCs cannot be unequivocally identified in the hematopoietic cell pool. Despite innumerable studies over the years, which focused on the search of the ideal phenotypic marker to selectively isolate stem cells, most of the known markers still define heterogeneous populations in different stages of commitment. Functional features attributed to stem cells have also been investigated, and among these the use of fluorescent markers which allow tracking of the cell division record of each cell. A second issue, after the initial isolation process, is the expansion ex vivo in order to obtain production of large numbers of homogeneous cell populations for both biological studies and clinical applications. Expansion ex vivo is difficult to modulate and normally occurs only along with commitment and consequent loss of multipotentiality. Moreover expansion obtained ex vivo is significantly reduced to that achievable in vivo. One of the key features of HSCs is a very slow proliferation rate, but when the appropriate stimuli are delivered, the proliferation rate can drastically increase. In normal physiological conditions a strict balance is maintained between the number of cells that maintain the original pool and those that proliferate and differentiate. Numerous data in recent years are providing some clue to elucidate the key steps in this tightly controlled process, but the dynamics that regulate which and how many cells self-renew to maintain the pool, and which proliferate and become committed to give rise to the mature blood elements, are still unclear.


Asunto(s)
Ciclo Celular/fisiología , Células Madre Hematopoyéticas/citología , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/fisiología , Citocinas/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Modelos Biológicos
17.
Leuk Lymphoma ; 43(1): 51-7, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11908736

RESUMEN

Homeostasis of the hematopoietic system is maintained by proliferation and differentiation of a small number of long-term surviving, self-renewing stem cells, which give rise to the fully mature elements. The fine interplay between differentiation, proliferation and death by apoptosis determines the equilibrium of this system. Thus, genes involved in the control of these processes are very important in the regulation and development of hematopoietic cells especially in the initial stages. The interactions among cyclins, their specific cyclin-dependent kinases (CDKs) and, a number of cyclin-dependent kinase inhibitors (CDKIs) such as p27 and p21, exert a direct control on the cell cycle but can also produce other independent effects on hematopoietic differentiation. Proteins of the Bcl-2 family are also crucial in regulating the balance between entry into apoptosis and survival capacity and their roles change in the course of differentiation. In addition, a number of autocrine and paracrine soluble factors (such as TGF-beta1) modulate the behavior and differentiation potential of hematopoietic elements. Studies on a few in vitro systems of early hematopoietic differentiation have stressed the importance of Bcl-2 and of the CDKIs p27 and p21 at this stage, have confirmed cell-cycle independent effects and have demonstrated how the modulation and the effects in response to different stimuli is mostly dependent on the differentiation stage of the target cells.


Asunto(s)
Ciclo Celular/fisiología , Hematopoyesis/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/genética , Ciclinas/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta1 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
18.
Oncotarget ; 5(8): 2052-64, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24903009

RESUMEN

Microenvironmental factors contribute to the immune dysfunction characterizing acute myeloid leukemia (AML). Indoleamine 2,3-dioxygenase 1 (IDO1) is an interferon (IFN)-γ-inducible enzyme that degrades tryptophan into kynurenine, which, in turn, inhibits effector T cells and promotes regulatory T-cell (Treg) differentiation. It is presently unknown whether childhood AML cells express IDO1 and whether IDO1 activity correlates with patient outcome. We investigated IDO1 expression and function in 37 children with newly diagnosed AML other than acute promyelocytic leukemia. Blast cells were cultured with exogenous IFN-γ for 24 hours, followed by the measurement of kynurenine production and tryptophan consumption. No constitutive expression of IDO1 protein was detected in blast cells from the 37 AML samples herein tested. Conversely, 19 out of 37 (51%) AML samples up-regulated functional IDO1 protein in response to IFN-γ. The inability to express IDO1 by the remaining 18 AML samples was not apparently due to a defective IFN-γ signaling circuitry, as suggested by the measurement of signal transducer and activator of transcription 3 (STAT3) phosphorylation. Co-immunoprecipitation assays indicated the occurrence of physical interactions between STAT3 and IDO1 in AML blasts. In line with this finding, STAT3 inhibitors abrogated IDO1 function in AML blasts. Interestingly, levels of IFN-γ were significantly higher in the bone marrow fluid of IDO-expressing compared with IDO-nonexpressing AMLs. In mixed tumor lymphocyte cultures (MTLC), IDO-expressing AML blasts blunted the ability of allogeneic naïve T cells to produce IFN-γ and promoted Treg differentiation. From a clinical perspective, the 8-year event-free survival was significantly worse in IDO-expressing children (16.4%, SE 9.8) as compared with IDO-nonexpressing ones (48.0%, SE 12.1; p=0.035). These data indicate that IDO1 expression by leukemia blasts negatively affects the prognosis of childhood AML. Moreover, they speak in favor of the hypothesis that IDO can be targeted, in adjunct to current chemotherapy approaches, to improve the clinical outcome of children with AML.


Asunto(s)
Biomarcadores de Tumor/análisis , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Leucemia Mieloide Aguda/enzimología , Adolescente , Western Blotting , Niño , Preescolar , Supervivencia sin Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Lactante , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
20.
Biomed Res Int ; 2013: 286368, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24063002

RESUMEN

Treg modulation has been hypothesized as one of the mechanisms by which antitumor necrosis factor α (TNFα) agents exert their action in rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). However, data in IBD are still conflicting. We evaluated CD4⁺CD25⁺FOXP3⁺ (Tregs) by flow cytometry in peripheral blood from 32 adult IBD patient before (T0) and after the induction of anti-TNFα therapy (T1). Eight healthy controls (HCs) were included. We also evaluated the number of FOXP3⁺ cells in the lamina propria (LP) in biopsies taken in a subset of patients and controls. Treg frequencies were significantly increased in peripheral blood from our patients after anti-TNFα therapy compared to T0. T1 but not T0 levels were higher than HC. The increase was detectable only in clinical responders to the treatment. A negative correlation was found among delta Treg levels and the age of patients or disease duration and with the activity score of Crohn's disease (CD). No significant differences were found in LP FOXP3⁺ cells. Our data suggest the possibility that in IBD patients the treatment with anti-TNFα may affect Treg percentages and that Treg modifications may correlate with clinical response, but differently in early versus late disease.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Factores de Transcripción Forkhead/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Colon/patología , Enfermedad de Crohn/sangre , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Enfermedades Inflamatorias del Intestino/sangre , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Membrana Mucosa/patología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA