Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(2): 53, 2023 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-36707427

RESUMEN

Chediak-Higashi syndrome (CHS) is a rare, autosomal recessive disorder caused by biallelic mutations in the lysosomal trafficking regulator (LYST) gene. Even though enlarged lysosomes and/or lysosome-related organelles (LROs) are the typical cellular hallmarks of CHS, they have not been investigated in human neuronal models. Moreover, how and why the loss of LYST function causes a lysosome phenotype in cells has not been elucidated. We report that the LYST-deficient human neuronal model exhibits lysosome depletion accompanied by hyperelongated tubules extruding from enlarged autolysosomes. These results have also been recapitulated in neurons differentiated from CHS patients' induced pluripotent stem cells (iPSCs), validating our model system. We propose that LYST ensures the correct fission/scission of the autolysosome tubules during autophagic lysosome reformation (ALR), a crucial process to restore the number of free lysosomes after autophagy. We further demonstrate that LYST is recruited to the lysosome membrane, likely to facilitate the fission of autolysosome tubules. Together, our results highlight the key role of LYST in maintaining lysosomal homeostasis following autophagy and suggest that ALR dysregulation is likely associated with the neurodegenerative CHS phenotype.


Asunto(s)
Síndrome de Chediak-Higashi , Proteínas de Transporte Vesicular , Humanos , Proteínas de Transporte Vesicular/genética , Lisosomas/fisiología , Orgánulos , Autofagia/fisiología , Síndrome de Chediak-Higashi/genética , Neuronas
2.
Blood ; 138(19): 1843-1854, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34046681

RESUMEN

Bruton tyrosine kinase inhibitors (BTKis) are a preferred treatment of patients with chronic lymphocytic leukemia (CLL). Indefinite therapy with BTKis, although effective, presents clinical challenges. Combination therapy can deepen responses, shorten treatment duration, and possibly prevent or overcome drug resistance. We previously reported on a CD19/CD3-bispecific antibody (bsAb) that recruits autologous T-cell cytotoxicity against CLL cells in vitro. Compared with observations with samples from treatment-naïve patients, T cells from patients being treated with ibrutinib expanded more rapidly and exerted superior cytotoxic activity in response to the bsAb. In addition to BTK, ibrutinib also inhibits interleukin-2 inducible T-cell kinase (ITK). In contrast, acalabrutinib, does not inhibit ITK. Whether ITK inhibition contributes to the observed immune effects is unknown. To better understand how BTKis modulate T-cell function and cytotoxic activity, we cultured peripheral blood mononuclear cells (PBMCs) from BTKi-naive and ibrutinib- or acalabrutinib-treated CLL patients with CD19/CD3 bsAb in vitro. T-cell expansion, activation, differentiation, and cytotoxicity were increased in PBMCs from patients on treatment with either BTKi compared with that observed for BKTi-naïve patients. BTKi therapy transcriptionally downregulated immunosuppressive effectors expressed by CLL cells, including cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and CD200. CTLA-4 blockade with ipilimumab in vitro increased the cytotoxic activity of the bsAb in BTKi-naïve but not BTKi-treated PBMCS. Taken together, BTKis enhance bsAb-induced cytotoxicity by relieving T cells of immunosuppressive restraints imposed by CLL cells. The benefit of combining bsAb immunotherapy with BTKis needs to be confirmed in clinical trials.


Asunto(s)
Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Piperidinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adenina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD19/inmunología , Benzamidas/uso terapéutico , Complejo CD3/inmunología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ipilimumab/uso terapéutico , Leucemia Linfocítica Crónica de Células B/inmunología , Masculino , Persona de Mediana Edad , Pirazinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
3.
Blood ; 132(5): 521-532, 2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-29743179

RESUMEN

The Bruton tyrosine kinase inhibitor ibrutinib induces high rates of clinical response in chronic lymphocytic leukemia (CLL). However, there remains a need for adjunct treatments to deepen response and to overcome drug resistance. Blinatumomab, a CD19/CD3 bispecific antibody (bsAb) designed in the BiTE (bispecific T-cell engager) format, is approved by the US Food and Drug Administration for the treatment of relapsed or refractory B-cell precursor acute lymphoblastic leukemia. Because of its short half-life of 2.1 hours, blinatumomab requires continuous intravenous dosing for efficacy. We developed a novel CD19/CD3 bsAb in the single-chain Fv-Fc format (CD19/CD3-scFv-Fc) with a half-life of ∼5 days. In in vitro experiments, both CD19/CD3-scFv-Fc and blinatumomab induced >90% killing of CLL cells from treatment-naïve patients. Antileukemic activity was associated with increased autologous CD8 and CD4 T-cell proliferation, activation, and granzyme B expression. In the NOD/SCID/IL2Rγnull patient-derived xenograft mouse model, once-weekly treatment with CD19/CD3-scFv-Fc eliminated >98% of treatment-naïve CLL cells in blood and spleen. By contrast, blinatumomab failed to induce a response, even when administered daily. We next explored the activity of CD19/CD3-scFv-Fc in the context of ibrutinib treatment and ibrutinib resistance. CD19/CD3-scFv-Fc induced more rapid killing of CLL cells from ibrutinib-treated patients than those from treatment-naïve patients. CD19/CD3-scFv-Fc also demonstrated potent activity against CLL cells from patients with acquired ibrutinib-resistance harboring BTK and/or PLCG2 mutations in vitro and in vivo using patient-derived xenograft models. Taken together, these data support investigation of CD19/CD3 bsAb's and other T cell-recruiting bsAb's as immunotherapies for CLL, especially in combination with ibrutinib or as rescue therapy in ibrutinib-resistant disease.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD19/inmunología , Complejo CD3/inmunología , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Pirazoles/farmacología , Pirimidinas/farmacología , Terapia Recuperativa , Adenina/análogos & derivados , Animales , Humanos , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Piperidinas , Anticuerpos de Cadena Única/inmunología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biol Blood Marrow Transplant ; 25(2): 216-222, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30292745

RESUMEN

Blockade of the T-cell exhaustion marker PD-1 to re-energize the immune response is emerging as a promising cancer treatment. Relapse of hematologic malignancy after allogeneic stem cell transplantation limits the success of this approach, and PD-1 blockade may hold therapeutic promise. However, PD-1 expression and its relationship with post-transplant relapse is poorly described. Because the donor immunity is activated by alloresponses, PD-1 expression may differ from nontransplanted individuals, and PD-1 blockade could risk graft-versus-host disease. Here we analyzed T-cell exhaustion marker kinetics and their relationship with leukemia relapse in 85 patients undergoing myeloablative T-cell-depleted HLA-matched stem cell transplantation. At a median follow-up of 3.5 years, 35 (44%) patients relapsed. PD-1 expression in CD4 and CD8 T cells was comparably elevated in relapsed and nonrelapsed cohorts. Helios+ regulatory T cells and CD8 effector memory cells at day 30 emerged as independent predictors of relapse. Although leukemia antigen-specific T cells did not overexpress PD-1, single-cell analysis revealed LAG3 and TIM3 overexpression at relapse. These findings indicate that PD-1 is an unreliable marker for leukemia-specific T-cell exhaustion in relapsing patients but implies other exhaustion markers and suppressor cells as relapse biomarkers.


Asunto(s)
Biomarcadores de Tumor/inmunología , Regulación Leucémica de la Expresión Génica/inmunología , Leucemia , Proteínas de Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Trasplante de Células Madre , Adulto , Aloinjertos , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Leucemia/inmunología , Leucemia/mortalidad , Leucemia/patología , Leucemia/terapia , Masculino , Persona de Mediana Edad , Recurrencia , Tasa de Supervivencia
5.
Blood ; 130(25): 2762-2773, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29030335

RESUMEN

Cancer cells frequently exhibit chromosomal abnormalities. Specific cytogenetic aberrations often are predictors of outcome, especially in hematologic neoplasms, such as monosomy 7 in myeloid malignancies. The functional consequences of aneuploidy at the cellular level are difficult to assess because of a lack of convenient markers to distinguish abnormal from diploid cells. We performed single-cell RNA sequencing (scRNA-seq) to study hematopoietic stem and progenitor cells from the bone marrow of 4 healthy donors and 5 patients with bone marrow failure and chromosome gain or loss. In total, transcriptome sequences were obtained from 391 control cells and 588 cells from patients. We characterized normal hematopoiesis as binary differentiation from stem cells to erythroid and myeloid-lymphoid pathways. Aneuploid cells were distinguished from diploid cells in patient samples by computational analyses of read fractions and gene expression of individual chromosomes. We confirmed assignment of aneuploidy to individual cells quantitatively, by copy-number variation, and qualitatively, by loss of heterozygosity. When we projected patients' single cells onto the map of normal hematopoiesis, diverse patterns were observed, broadly reflecting clinical phenotypes. Patients' monosomy 7 cells showed downregulation of genes involved in immune response and DNA damage checkpoint and apoptosis pathways, which may contribute to the clonal expansion of monosomy 7 cells with accumulated gene mutations. scRNA-seq is a powerful technique through which to infer the functional consequences of chromosome gain and loss and explore gene targets for directed therapy.


Asunto(s)
Aneuploidia , Células Madre Hematopoyéticas , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Adulto , Células de la Médula Ósea , Enfermedades de la Médula Ósea/genética , Enfermedades de la Médula Ósea/patología , Estudios de Casos y Controles , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 7 , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad
6.
Haematologica ; 104(5): 894-906, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30545929

RESUMEN

Long noncoding RNAs (lncRNAs) are regulators of cell differentiation and development. The lncRNA transcriptome in human hematopoietic stem and progenitor cells is not comprehensively defined. We investigated lncRNAs in 979 human bone marrow-derived CD34+ cells by single cell RNA sequencing followed by de novo transcriptome reconstruction. We identified 3,173 lncRNAs in total, among which 2,365 were previously unknown, and we characterized lncRNA stem, differentiation, and maturation signatures. lncRNA expression exhibited high cell-to-cell variation, which was only apparent in single cell analysis. lncRNA expression followed a lineage-specific and highly dynamic pattern during early hematopoiesis. lncRNAs in hematopoietic cells closely correlated with protein-coding genes of known functions in the regulation of hematopoiesis and cell fate decisions, and the potential regulatory roles of lncRNAs in hematopoiesis were imputed by projection from protein-coding genes with a "guilt-by-association" approach. We characterized lncRNAs preferentially expressed in hematopoietic stem cells and in various downstream differentiated lineage progenitors. We also profiled lncRNA expression in single cells from patients with myelodysplastic syndromes and in aneuploid cells in particular. Our study provides a global view of lncRNAs in human hematopoietic stem and progenitor cells. We observed a highly ordered pattern of lncRNA expression and participation in regulation of early hematopoiesis, and coordinate aberrant messenger RNA and lncRNA transcriptomes in dysplastic hematopoiesis. (Registered at clinicaltrials.gov with identifiers: 00001620, 00001397).


Asunto(s)
Biomarcadores de Tumor/genética , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Síndromes Mielodisplásicos/genética , ARN Largo no Codificante/genética , Análisis de la Célula Individual/métodos , Transcriptoma , Médula Ósea/patología , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/citología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndromes Mielodisplásicos/patología
7.
J Immunol ; 199(2): 477-488, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28630090

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired disorder originating from hematopoietic stem cells and is a life-threating disease characterized by intravascular hemolysis, bone marrow (BM) failure, and venous thrombosis. The etiology of PNH is a somatic mutation in the phosphatidylinositol glycan class A gene (PIG-A) on the X chromosome, which blocks synthesis of the glycolipid moiety and causes deficiency in GPI-anchored proteins. PNH is closely related to aplastic anemia, in which T cells mediate destruction of BM. To identify aberrant molecular mechanisms involved in immune targeting of hematopoietic stem cells in BM, we applied RNA-seq to examine the transcriptome of T cell subsets (CD4+ naive, CD4+ memory, CD8+ naive, and CD8+ memory) from PNH patients and healthy control subjects. Differentially expressed gene analysis in four different T cell subsets from PNH and healthy control subjects showed distinct transcriptional profiles, depending on the T cell subsets. By pathway analysis, we identified novel signaling pathways in T cell subsets from PNH, including increased gene expression involved in TNFR, IGF1, NOTCH, AP-1, and ATF2 pathways. Dysregulation of several candidate genes (JUN, TNFAIP3, TOB1, GIMAP4, GIMAP6, TRMT112, NR4A2, CD69, and TNFSF8) was validated by quantitative real-time RT-PCR and flow cytometry. We have demonstrated molecular signatures associated with positive and negative regulators in T cells, suggesting novel pathophysiologic mechanisms in PNH. These pathways may be targets for new strategies to modulate T cell immune responses in BM failure.


Asunto(s)
Hemoglobinuria Paroxística/inmunología , Redes y Vías Metabólicas/genética , Subgrupos de Linfocitos T/inmunología , Transcriptoma , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Adulto , Ligando CD30/genética , Ligando CD30/metabolismo , Linfocitos T CD4-Positivos/inmunología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/fisiopatología , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Metiltransferasas/genética , Metiltransferasas/metabolismo , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Transducción de Señal/genética , Subgrupos de Linfocitos T/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
9.
Biol Blood Marrow Transplant ; 24(3): 460-466, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29197677

RESUMEN

Various approaches have been developed for ex vivo T cell depletion in allogeneic stem cell transplantation to prevent graft-versus-host disease (GVHD). Direct comparisons of T cell depletion strategies have not been well studied, however. We evaluated cellular and plasma biomarkers in 2 different graft manipulation strategies, CD3+CD19+ cell depletion (CD3/19D) versus CD34+ selection (CD34S), and their associations with clinical outcomes. Identical conditions, including the myeloablative preparative regimen, HLA-identical sibling donor, GVHD prophylaxis, and graft source, were used in the 2 cohorts. Major clinical outcomes were similar in the 2 groups in terms of overall survival, nonrelapse mortality, and cumulative incidence of relapse; however, the cumulative incidence of acute GVHD trended to be higher in the CD3/19D cohort compared with the CD34S cohort. A distinct biomarker profile was noted in the CD3/19D cohort: higher levels of ST2, impaired Helios- FoxP3+Treg reconstitution, and rapid reconstitution of naïve, Th2, and Th17 CD4 cells in the early post-transplantation period. In vitro graft replication studies confirmed that CD3/19D disproportionately depleted Tregs and other CD4 subset repertoires in the graft. This study confirms the utility of biomarker monitoring, which can be directly correlated with biological consequences and possible future therapeutic indications.


Asunto(s)
Antígenos CD19/inmunología , Antígenos CD34/inmunología , Neoplasias Hematológicas , Depleción Linfocítica , Trasplante de Células Madre de Sangre Periférica , Hermanos , Linfocitos T Colaboradores-Inductores/inmunología , Donantes de Tejidos , Acondicionamiento Pretrasplante/métodos , Adolescente , Adulto , Anciano , Aloinjertos , Niño , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/terapia , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
10.
Blood Cells Mol Dis ; 69: 10-22, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29324392

RESUMEN

DNA methyltransferase 3A (DNMT3A) mediates de novo DNA methylation. Mutations in DNMT3A are associated with hematological malignancies, most frequently acute myeloid leukemia. DNMT3A mutations are hypothesized to establish a pre-leukemic state, rendering cells vulnerable to secondary oncogenic mutations and malignant transformation. However, the mechanisms by which DNMT3A mutations contribute to leukemogenesis are not well-defined. Here, we successfully created four DNMT3A-mutated K562 cell lines with frameshift mutations resulting in truncated DNMT3A proteins. DNMT3A-mutated cell lines exhibited significantly impaired growth and increased apoptotic activity compared to wild-type (WT) cells. Consistent with previous studies, DNMT3A-mutated cells displayed impaired differentiation capacity. RNA-seq was used to compare transcriptomes of DNMT3A-mutated and WT cells; DNMT3A ablation resulted in downregulation of genes involved in spliceosome function, causing dysfunction of RNA splicing. Unexpectedly, we observed DNMT3A-mutated cells to exhibit marked genomic instability and an impaired DNA damage response compared to WT. CRISPR/Cas9-mediated DNMT3A-mutated K562 cells may be used to model effects of DNMT3A mutations in human cells. Our findings implicate aberrant splicing and induction of genomic instability as potential mechanisms by which DNMT3A mutations might predispose to malignancy.


Asunto(s)
Sistemas CRISPR-Cas , ADN (Citosina-5-)-Metiltransferasas/genética , Edición Génica , Inestabilidad Genómica , Empalme del ARN , Apoptosis/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Daño del ADN , ADN Metiltransferasa 3A , Humanos , Células K562 , Mutación , Análisis de Secuencia de ADN , Empalmosomas/metabolismo
11.
J Immunol ; 196(4): 1568-78, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26764034

RESUMEN

Memory stem T cells (TSCMs) constitute a long-lived, self-renewing lymphocyte population essential for the maintenance of functional immunity. Hallmarks of autoimmune disease pathogenesis are abnormal CD4(+) and CD8(+) T cell activation. We investigated the TSCM subset in 55, 34, 43, and 5 patients with acquired aplastic anemia (AA), autoimmune uveitis, systemic lupus erythematosus, and sickle cell disease, respectively, as well as in 41 age-matched healthy controls. CD8(+) TSCM frequency was significantly increased in AA compared with healthy controls. An increased CD8(+) TSCM frequency at diagnosis was associated with responsiveness to immunosuppressive therapy, and an elevated CD8(+) TSCM population after immunosuppressive therapy correlated with treatment failure or relapse in AA patients. IFN-γ and IL-2 production was significantly increased in various CD8(+) and CD4(+) T cell subsets in AA patients, including CD8(+) and CD4(+) TSCMs. CD8(+) TSCM frequency was also increased in patients with autoimmune uveitis or sickle cell disease. A positive correlation between CD4(+) and CD8(+) TSCM frequencies was found in AA, autoimmune uveitis, and systemic lupus erythematosus. Evaluation of PD-1, CD160, and CD244 expression revealed that TSCMs were less exhausted compared with other types of memory T cells. Our results suggest that the CD8(+) TSCM subset is a novel biomarker and a potential therapeutic target for AA.


Asunto(s)
Anemia Aplásica/inmunología , Anemia Aplásica/terapia , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Células Precursoras de Linfocitos T/inmunología , Adulto , Anciano , Anemia Aplásica/sangre , Anemia Aplásica/diagnóstico , Anemia de Células Falciformes/diagnóstico , Anemia de Células Falciformes/inmunología , Enfermedades Autoinmunes/inmunología , Biomarcadores/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/clasificación , Femenino , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/inmunología , Activación de Linfocitos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Recurrencia , Subgrupos de Linfocitos T , Insuficiencia del Tratamiento , Uveítis/diagnóstico , Uveítis/inmunología
12.
Br J Haematol ; 177(1): 136-141, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28151558

RESUMEN

The aetiology of paroxysmal nocturnal haemoglobinuria (PNH) is a somatic mutation in the X-linked phosphatidylinositol glycan class A gene (PIGA), resulting in global deficiency of glycosyl phosphatidylinositol-anchored proteins (GPI-APs). This study applied RNA-sequencing to examine functional effects of the PIGA mutation in human granulocytes. CXCR2 expression was increased in GPI-AP- compared to GPI-AP+ granulocytes. Macrophage migration inhibitory factor, a CXCR2 agonist, was significantly higher in plasma of PNH patients. Nuclear factor-κB phosphorylation was upregulated in GPI-AP- compared with GPI-AP+ granulocytes. Our data suggest novel mechanisms in PNH, not obviously predicted by decreased production of the GPI moiety.


Asunto(s)
Regulación de la Expresión Génica , Granulocitos/metabolismo , Hemoglobinuria Paroxística/genética , Receptores de Interleucina-8B/genética , Transcriptoma , Biomarcadores , Estudios de Casos y Controles , Análisis por Conglomerados , Citometría de Flujo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
13.
Blood ; 126(24): 2621-31, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26491068

RESUMEN

Interferon gamma (IFN-γ) has been reported to have both negative and positive activity on hematopoietic cells, adding complexity to the interpretation of its pleiotropic functions. We examined the effects of IFN-γ on murine hematopoietic stem cells (HSCs) and progenitors in vitro and in vivo by using mouse models. IFN-γ treatment expanded bone marrow (BM) c-Kit(+)Sca1(+)Lin(-) (KSL) cell number but reduced BM KLCD150(+) and KLCD150(+)CD48(-) cells. IFN-γ-expanded KSL cells engrafted poorly when tested by competitive repopulation in vivo. KSL, KLCD150(+), and KLCD150(+)CD48(-) cells from IFN-γ-treated animals all showed significant upregulation in Fas expression. When cocultured with activated T cells in vitro, KSL and KLCD150(+) cells from IFN-γ-treated donors showed increased apoptosis relative to those from untreated animals, and infusion of activated CD8 T cells into IFN-γ-injected animals in vivo led to partial elimination of KSL cells. Exposure of BM cells or KSL cells to IFN-γ increased expression of Fas, caspases, and related proapoptotic genes and decreased expression of Ets-1 and other hematopoietic genes. In mouse models of BM failure, mice genetically deficient in IFN-γ receptor expression showed attenuation of immune-mediated marrow destruction, whereas effector lymphocytes from IFN-γ-deficient donors were much less potent in initiating BM damage. We conclude that the activity of IFN-γ on murine hematopoiesis is context dependent. IFN-γ-augmented apoptotic gene expression facilitates destruction of HSCs and progenitors in the presence of activated cytotoxic T cells, as occurs in human BM failure.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Hemoglobinuria Paroxística/inmunología , Interferón gamma/farmacología , Linfocitos T Citotóxicos/inmunología , Anemia Aplásica , Animales , Apoptosis/efectos de los fármacos , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Trasplante de Médula Ósea , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Modelos Animales de Enfermedad , Proteína Ligando Fas/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Hemoglobinuria Paroxística/fisiopatología , Interferón gamma/fisiología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Receptores de Interferón/deficiencia , Receptores de Interferón/fisiología , Receptor fas/biosíntesis , Receptor fas/genética , Receptor de Interferón gamma
14.
Haematologica ; 102(10): 1691-1703, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28729300

RESUMEN

Acquired aplastic anemia, the prototypical bone marrow failure disease, is characterized by pancytopenia and marrow hypoplasia. Most aplastic anemia patients respond to immunosuppressive therapy, usually with anti-thymocyte globulin and cyclosporine, but some relapse on cyclosporine withdrawal or require long-term administration of cyclosporine to maintain blood counts. In this study, we tested efficacy of rapamycin as a new or alternative treatment in mouse models of immune-mediated bone marrow failure. Rapamycin ameliorated pancytopenia, improved bone marrow cellularity, and extended animal survival in a manner comparable to the standard dose of cyclosporine. Rapamycin effectively reduced Th1 inflammatory cytokines interferon-γ and tumor necrosis factor-α, increased the Th2 cytokine interleukin-10, stimulated expansion of functional regulatory T cells, eliminated effector CD8+ T cells (notably T cells specific to target cells bearing minor histocompatibility antigen H60), and preserved hematopoietic stem and progenitor cells. Rapamycin, but not cyclosporine, reduced the proportion of memory and effector T cells and maintained a pool of naïve T cells. Cyclosporine increased cytoplasmic nuclear factor of activated T-cells-1 following T-cell receptor stimulation, whereas rapamycin suppressed phosphorylation of two key signaling molecules in the mammalian target of rapamycin pathway, S6 kinase and protein kinase B. In summary, rapamycin was an effective therapy in mouse models of immune-mediated bone marrow failure, acting through different mechanisms to cyclosporine. Its specific expansion of regulatory T cells and elimination of clonogenic CD8+ effectors support its potential clinical utility in the treatment of aplastic anemia.


Asunto(s)
Anemia Aplásica/inmunología , Anemia Aplásica/patología , Enfermedades de la Médula Ósea/inmunología , Enfermedades de la Médula Ósea/patología , Médula Ósea/inmunología , Médula Ósea/patología , Hemoglobinuria Paroxística/inmunología , Hemoglobinuria Paroxística/patología , Inmunosupresores/farmacología , Sirolimus/farmacología , Anemia Aplásica/tratamiento farmacológico , Anemia Aplásica/metabolismo , Anemia Aplásica/mortalidad , Animales , Médula Ósea/efectos de los fármacos , Enfermedades de la Médula Ósea/tratamiento farmacológico , Enfermedades de la Médula Ósea/mortalidad , Trastornos de Fallo de la Médula Ósea , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/mortalidad , Memoria Inmunológica , Ratones , Pancitopenia/inmunología , Pancitopenia/patología , Transducción de Señal , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Resultado del Tratamiento
15.
Mol Ther ; 24(9): 1655-64, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27401140

RESUMEN

Selective depletion (SD) of alloreactive T cells from allogeneic hematopoeitic stem cell transplants to prevent graft-versus-host disease (GVHD) without compromising immune reconstitution and antitumor responses remains a challenge. Here, we demonstrate a novel SD strategy whereby alloreacting T cells are efficiently deleted ex vivo with adenosine. SD was achieved in human leukocyte antigen (HLA) mismatched cocultures by multiple exposures to 2 mmol/l adenosine over 7 days. Adenosine depleted greater than to 90% of alloproliferating T cells in mismatched, haploidentical, and matched sibling pairs while conserving response to third-party antigens. Alloreactive CD4 and CD8 T cells were targeted for depletion while NK and B cells were preserved. Our novel approach also preserved nonalloreactive naive, central, and effector memory T-cell subsets, Tregs, and notably preserved T-cell responses against DNA viruses that contribute to transplant related mortality after allogeneic hematopoeitic stem cell transplants. Additionally, T cells recognizing leukemia-associated antigens were efficiently generated in vitro from the cell product post-SD. This study is the first to demonstrate that adenosine depletion of alloactivated T cells maintains a complete immune cell profile and recall viral responses. Expansion of tumor antigen-specific subsets postdepletion opens the possibility of generating T-cell products capable of graft-versus-tumor responses without causing GVHD.


Asunto(s)
Adenosina/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Inmunidad , Leucemia/inmunología , Depleción Linfocítica , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Virus/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias , Supervivencia Celular/efectos de los fármacos , Enfermedad Injerto contra Huésped/etiología , Haplotipos , Humanos , Leucemia/complicaciones , Leucemia/terapia , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Persona de Mediana Edad , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/metabolismo , Donantes de Tejidos , Adulto Joven
16.
Br J Haematol ; 175(3): 427-439, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27433923

RESUMEN

Although recent observations implicate the importance of telomerase activity in acute myeloid leukaemia (AML), the roles of epigenetic regulations of the TERT gene in leukaemogenesis, drug resistance and clinical prognosis in AML are not fully understood. We developed a quantitative pyrosequencing-based methylation assay covering the TERT proximal promoter and a partial exon 1 (TERTpro/Ex1) region and tested both cell lines and primary leukaemia cells derived from AML and AML with preceding myelodysplastic syndrome (AML/MDS) patients (n = 43). Prognostic impact of methylation status of the upstream TERT promoter region was assessed by the Kaplan-Meier method. The activity of the telomerase inhibitor, imetelstat, was measured using leukaemia cell lines. The TERTpro/Ex1 region was highly methylated in all cell lines and primary leukaemia cells showed diverse methylation profiles. Most cases showed hypermethylated regions at the upstream TERTpro/Ex1 region, which were associated with inferior patient survival. TERTpro/Ex1 methylation status was correlated with the cytotoxicity to imetelstat and its combination with hypomethylating agent enhanced the cytotoxicity of imetelstat. AML cell lines and primary blasts harbour distinct TERTpro/Ex1 methylation profiles that could serve as a prognostic biomarker of AML. However, validation in a large cohort of patients is necessary to confirm our findings.


Asunto(s)
Epigénesis Genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Regiones Promotoras Genéticas , Telomerasa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Línea Celular , Análisis por Conglomerados , Islas de CpG , Metilación de ADN , Resistencia a Antineoplásicos/genética , Exones , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Inmunofenotipificación , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/mortalidad , Pronóstico , Telomerasa/antagonistas & inhibidores , Homeostasis del Telómero , Sitio de Iniciación de la Transcripción , Resultado del Tratamiento , Adulto Joven
17.
Haematologica ; 101(1): 57-67, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26589913

RESUMEN

Acquired aplastic anemia is an immune-mediated disease, in which T cells target hematopoietic cells; at presentation, the bone marrow is replaced by fat. It was reported that bone marrow adipocytes were negative regulators of hematopoietic microenvironment. To examine the role of adipocytes in bone marrow failure, we investigated peroxisomal proliferator-activated receptor gamma, a key transcription factor in adipogenesis, utilizing an antagonist of this factor called bisphenol-A-diglycidyl-ether. While bisphenol-A-diglycidyl-ether inhibited adipogenesis as expected, it also suppressed T cell infiltration of bone marrow, reduced plasma inflammatory cytokines, decreased expression of multiple inflammasome genes, and ameliorated marrow failure. In vitro, bisphenol-A-diglycidyl-ether suppressed activation and proliferation, and reduced phospholipase C gamma 1 and nuclear factor of activated T-cells 1 expression, as well as inhibiting calcium flux in T cells. The in vivo effect of bisphenol-A-diglycidyl-ether on T cells was confirmed in a second immune-mediated bone marrow failure model, using different strains and non-major histocompatibility antigen mismatched: bisphenol-A-diglycidyl-ether ameliorated marrow failure by inhibition of T cell infiltration of bone marrow. Our data indicate that peroxisomal proliferator-activated receptor gamma antagonists may attenuate murine immune-mediated bone marrow failure, at least in part, by suppression of T cell activation, which might hold implications in the application of peroxisomal proliferator-activated receptor gamma antagonists in immune-mediated pathophysiologies, both in the laboratory and in the clinic. Genetically "fatless" mice developed bone marrow failure with accumulation of marrow adipocytes in our model, even in the absence of body fat, suggesting different mechanisms of systematic and marrow adipogenesis and physiologic versus pathophysiologic fat accumulation.


Asunto(s)
Anemia Aplásica/inmunología , Compuestos de Bencidrilo/farmacología , Médula Ósea/inmunología , Compuestos Epoxi/farmacología , Inmunidad Celular/efectos de los fármacos , PPAR gamma/antagonistas & inhibidores , Linfocitos T/inmunología , Anemia Aplásica/patología , Animales , Médula Ósea/patología , Ratones , PPAR gamma/inmunología , Linfocitos T/patología
18.
Stem Cells ; 33(1): 91-100, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25330775

RESUMEN

The high risk of insertional oncogenesis reported in clinical trials using integrating retroviral vectors to genetically modify hematopoietic stem and progenitor cells (HSPCs) requires the development of safety strategies to minimize risks associated with novel cell and gene therapies. The ability to ablate genetically modified cells in vivo is desirable, should an abnormal clone emerge. Inclusion of "suicide genes" in vectors to facilitate targeted ablation of vector-containing abnormal clones in vivo is one potential safety approach. We tested whether the inclusion of the "inducible Caspase-9" (iCasp9) suicide gene in a gamma-retroviral vector facilitated efficient elimination of vector-containing HSPCs and their hematopoietic progeny in vivo long-term, in an autologous non-human primate transplantation model. Following stable engraftment of iCasp9 expressing hematopoietic cells in rhesus macaques, administration of AP1903, a chemical inducer of dimerization able to activate iCasp9, specifically eliminated vector-containing cells in all hematopoietic lineages long-term, suggesting activity at the HSPC level. Between 75% and 94% of vector-containing cells were eliminated by well-tolerated AP1903 dosing, but lack of complete ablation was linked to lower iCasp9 expression in residual cells. Further investigation of resistance mechanisms demonstrated upregulation of Bcl-2 in hematopoietic cell lines transduced with the vector and resistant to AP1903 ablation. These results demonstrate both the potential and the limitations of safety approaches using iCasp9 to HSPC-targeted gene therapy settings, in a model with great relevance to clinical development.


Asunto(s)
Caspasa 9/genética , Genes Transgénicos Suicidas , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Animales , Apoptosis/fisiología , Caspasa 9/biosíntesis , Terapia Genética , Vectores Genéticos , Células Madre Hematopoyéticas/enzimología , Macaca mulatta
19.
Haematologica ; 100(12): 1534-45, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26354756

RESUMEN

Emerging evidence indicates that microRNA control and modulate immunity. MicroRNA have not been investigated in acquired aplastic anemia, a T-cell-mediated immune disease. Analysis of 84 microRNA expression levels in CD4(+) and CD8(+) T cells of patients with aplastic anemia revealed concurrent down-regulation of miR-126-3p, miR-145-5p, miR-223-3p, and miR-199a-5p (>3-fold change, P<0.05) in both T-cell populations, which were unique in aplastic anemia compared to other hematologic disorders. MiR-126-3p and miR-223-3p were down-regulated in CD4(+) T effector memory cells, and miR-126-3p, miR-145-5p, and miR-223-3p were down-regulated in CD8(+) T effector memory and terminal effector cells. Successful immunosuppressive therapy was associated with restoration to normal expression levels of miR-126-3p, miR-145-5p, and miR-223-3p (>2-fold change, P<0.05). In CD4(+) and CD8(+) T cells in aplastic anemia patients, MYC and PIK3R2 were up-regulated and proved to be targets of miR-145-5p and miR-126-3p, respectively. MiR-126-3p and miR-145-5p knockdown promoted proliferation and increased interferon-γ and granzyme B production in both CD4(+) and CD8(+) T cells. Our work describes previously unknown regulatory roles of microRNA in T-cell activation in aplastic anemia, which may open a new perspective for development of effective therapy. Clinicaltrials.gov identifier: NCT 01623167.


Asunto(s)
Anemia Aplásica/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , MicroARNs/inmunología , Adolescente , Adulto , Anciano , Anemia Aplásica/patología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Femenino , Humanos , Activación de Linfocitos , Masculino , Persona de Mediana Edad
20.
Cytotherapy ; 17(12): 1675-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26422657

RESUMEN

BACKGROUND AIMS: With the increasing use of cell therapies involving immune modulatory cells, there is a need for a simple standardized method to evaluate and compare the suppressive potency of different cell products. We used the Karpas 299 (K299) cell line as the reference suppressor cell to develop a standardized suppression assay to quantify the immune-modulatory capacity of bone marrow-derived mesenchymal stromal cells (BM-MSCs). METHODS: Healthy donor CD4 T cells were co-cultured with the K299 cell line or with third-party BM-MSCs. After stimulation with anti-CD3/CD28 beads, CD154 activation and proliferation of CD4 T cells were measured to calculate suppression. RESULTS: The K299 cell line reproducibly suppressed both the activation and proliferation of healthy donor CD4 T cells in a dose-dependent manner. A rapid (16-h) assay that was based on activation-suppression was selected for development. In replicate testing, there was an inherent variability of suppression of 11% coefficient of variation between different responder T cells. Suppression by BM-MSCs on different responders correlated with suppression by K299. We therefore used K299 suppression as the reference to define suppression potency of BM-MSCs in K299 Suppression Units. We found that inter-donor variability, passage number, method of manufacture and exposure of BM-MSCs to steroids or interferon-γ all affected BM-MSC potency of suppression. CONCLUSIONS: This method provides a platform for standardizing suppressor function to facilitate comparisons between laboratories and for use as a cell product release assay.


Asunto(s)
Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/citología , Terapia de Inmunosupresión/métodos , Interferón gamma/farmacología , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/citología , Anticuerpos/inmunología , Anticuerpos/farmacología , Bioensayo , Antígenos CD28/inmunología , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/metabolismo , Línea Celular , Proliferación Celular , Supervivencia Celular/inmunología , Técnicas de Cocultivo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA