Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
2.
Sci Rep ; 11(1): 13958, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34230565

RESUMEN

Although the physiological function of the omentum remains elusive, it has been proposed that it plays an important role in fat storage, immune regulation, and regeneration of injured tissues and organs. Although the omentum undergoes expansion upon activation, reports on the accurate assessment of increased cell types and the origin of the increased cells remain limited. To investigate this aspect, the omenta of parabiotic mice were subjected to activation using distinct fluorescent markers and single-cell RNA sequencing (scRNA-seq) was performed to identify circulation-derived omental cells. We found that a considerable number of circulating cells contributed to the activation of the omentum. The omental cells derived from circulating cells exhibited morphological features similar to those of fibroblasts. scRNA-seq revealed the existence of a novel cell population that co-expressed macrophage and fibroblast markers in the activated omentum, suggesting that it corresponded to circulating macrophage-derived fibroblast-like cells. Lineage tracing experiments revealed that most fibroblasts in the activated omentum were not derived from WT1-positive mesenchymal progenitors. The cell cluster also expressed various chemokine genes, indicating its role in the activation and recruitment of immune cells. These results provide important information regarding the activation of the omentum.


Asunto(s)
Modelos Biológicos , Epiplón/patología , Parabiosis , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Adipocitos/patología , Animales , Fibroblastos/patología , Ratones Endogámicos C57BL , Microscopía Confocal , ARN no Traducido/genética , ARN no Traducido/metabolismo , Células Madre/patología
3.
Brain Commun ; 2(1): fcz048, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32954314

RESUMEN

Accumulated experience supports the efficacy of allogenic haematopoietic stem cell transplantation in arresting the progression of childhood-onset cerebral form of adrenoleukodystrophy in early stages. For adulthood-onset cerebral form of adrenoleukodystrophy, however, there have been only a few reports on haematopoietic stem cell transplantation and the clinical efficacy and safety of that for adulthood-onset cerebral form of adrenoleukodystrophy remain to be established. To evaluate the clinical efficacy and safety of haematopoietic stem cell transplantation, we conducted haematopoietic stem cell transplantation on 12 patients with adolescent-/adult-onset cerebral form/cerebello-brainstem form of adrenoleukodystrophy in a single-institution-based prospective study. Through careful prospective follow-up of 45 male adrenoleukodystrophy patients, we aimed to enrol patients with adolescent-/adult-onset cerebral form/cerebello-brainstem form of adrenoleukodystrophy at early stages. Indications for haematopoietic stem cell transplantation included cerebral form of adrenoleukodystrophy or cerebello-brainstem form of adrenoleukodystrophy with Loes scores up to 13, the presence of progressively enlarging white matter lesions and/or lesions with gadolinium enhancement on brain MRI. Clinical outcomes of haematopoietic stem cell transplantation were evaluated by the survival rate as well as by serial evaluation of clinical rating scale scores and neurological and MRI findings. Clinical courses of eight patients who did not undergo haematopoietic stem cell transplantation were also evaluated for comparison of the survival rate. All the patients who underwent haematopoietic stem cell transplantation survived to date with a median follow-up period of 28.6 months (4.2-125.3 months) without fatality. Neurological findings attributable to cerebral/cerebellar/brainstem lesions became stable or partially improved in all the patients. Gadolinium-enhanced brain lesions disappeared or became obscure within 3.5 months and the white matter lesions of MRI became stable or small. The median Loes scores before haematopoietic stem cell transplantation and at the last follow-up visit were 6.0 and 5.25, respectively. Of the eight patients who did not undergo haematopoietic stem cell transplantation, six patients died 69.1 months (median period; range 16.0-104.1 months) after the onset of the cerebral/cerebellar/brainstem lesions, confirming that the survival probability was significantly higher in patients with haematopoietic stem cell transplantation compared with that in patients without haematopoietic stem cell transplantation (P = 0.0089). The present study showed that haematopoietic stem cell transplantation was conducted safely and arrested the inflammatory demyelination in all the patients with adolescent-/adult-onset cerebral form/cerebello-brainstem form of adrenoleukodystrophy when haematopoietic stem cell transplantation was conducted in the early stages. Further studies are warranted to optimize the procedures of haematopoietic stem cell transplantation for adolescent-/adult-onset cerebral form/cerebello-brainstem form of adrenoleukodystrophy.

4.
Sci Rep ; 8(1): 15855, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30367142

RESUMEN

Chronic myelomonocytic leukemia (CMML) is an entity of myelodysplastic syndrome/myeloproliferative neoplasm. Although CMML can be cured with allogeneic stem cell transplantation, its prognosis is generally very poor due to the limited efficacy of chemotherapy and to the patient's age, which is usually not eligible for transplantation. Comprehensive analysis of CMML pathophysiology and the development of therapeutic agents have been limited partly due to the lack of cell lines in CMML and the limited developments of mouse models. After successfully establishing patient's derived disease-specific induced pluripotent stem cells (iPSCs) derived from a patient with CMML, we utilized these CMML-iPSCs to achieve hematopoietic re-differentiation in vitro, created a humanized CMML mouse model via teratomas, and developed a drug-testing system. The clinical characteristics of CMML were recapitulated following hematopoietic re-differentiation in vitro and a humanized CMML mouse model in vivo. The drug-testing system using CMML-iPSCs identified a MEK inhibitor, a Ras inhibitor, and liposomal clodronate as potential drugs for treating CMML. Clodronate is a drug commonly used as a bisphosphonate for osteoporosis. In this study, the liposomalization of clodronate enhanced its effectiveness in these assays, suggesting that this variation of clodronate may be adopted as a repositioned drug for CMML therapy.


Asunto(s)
Ácido Clodrónico/uso terapéutico , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Liposomas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Anciano , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Leucemia Mielomonocítica Crónica/patología , Leucemia Mielomonocítica Crónica/terapia , Masculino , Ratones , Neurofibromina 1/metabolismo , Fosforilación , Factor de Transcripción STAT5/metabolismo , Teratoma/metabolismo , Teratoma/patología , Trasplante Heterólogo
5.
Stem Cell Reports ; 10(3): 1115-1130, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29429960

RESUMEN

Properties of cancer stem cells involved in drug resistance and relapse have significant effects on clinical outcome. Although tyrosine kinase inhibitors (TKIs) have dramatically improved survival of patients with chronic myeloid leukemia (CML), TKIs have not fully cured CML due to TKI-resistant CML stem cells. Moreover, relapse after discontinuation of TKIs has not been predicted in CML patients with the best TKI response. In our study, a model of CML stem cells derived from CML induced pluripotent stem cells identified ADAM8 as an antigen of TKI-resistant CML cells. The inhibition of expression or metalloproteinase activity of ADAM8 restored TKI sensitivity in primary samples. In addition, residual CML cells in patients with optimal TKI response were concentrated in the ADAM8+ population. Our study demonstrates that ADAM8 is a marker of residual CML cells even in patients with optimal TKI response and would be a predictor of relapse and a therapeutic target of TKI-resistant CML cells.


Asunto(s)
Proteínas ADAM/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas de la Membrana/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células K562 , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo
6.
Sci Rep ; 7(1): 9891, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860595

RESUMEN

The murine intestine, like that of other mammalians, continues to develop after birth until weaning; however, whether this occurs in response to an intrinsic developmental program or food intake remains unclear. Here, we report a novel system for the allotransplantation of small intestine and colon harvested from Lgr5 EGFP-IRES-CreERT2/+; Rosa26 rbw/+ mice immediately after birth into the subrenal capsule of wild-type mice. By histological and immunohistochemical analysis, the developmental process of transplanted small intestine and colon was shown to be comparable with that of the native tissues: mature intestines equipped with all cell types were formed, indicating that these organs do not require food intake for development. The intestinal stem cells in transplanted tissues were shown to self-renew and produce progeny, resulting in the descendants of the stem cells occupying the crypt-villus unit of the small intestine or the whole crypt of the colon. Collectively, these findings indicate that neonatal intestine development follows an intrinsic program even in the absence of food stimuli.


Asunto(s)
Diferenciación Celular , Colon/citología , Colon/fisiología , Intestino Delgado/citología , Intestino Delgado/fisiología , Células Madre/citología , Células Madre/metabolismo , Aloinjertos , Animales , Animales Recién Nacidos , Biomarcadores , Proliferación Celular , Digestión , Técnica del Anticuerpo Fluorescente , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones
7.
Sci Rep ; 7: 41838, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176811

RESUMEN

Although the existence of cancer stem cells in intestine tumors has been suggested, direct evidence has not been yet provided. Here, we showed, using the multicolor lineage-tracing method and mouse models of intestinal adenocarcinoma and adenoma that Bmi1- or Lgr5- positive tumorigenic cells clonally expanded in proliferating tumors. At tumor initiation and during tumor propagation in the colon, the descendants of Lgr5-positive cells clonally proliferated to form clusters. Clonal analysis using ubiquitous multicolor lineage tracing revealed that colon tumors derived from Lgr5-positive cells were monoclonal in origin but eventually merged with neighboring tumors, producing polyclonal tumors at the later stage. In contrast, the origin of small intestine tumors was likely polyclonal, and during cancer progression some clones were eliminated, resulting in the formation of monoclonal tumors, which could merge similar to colon tumors. These results suggest that in proliferating intestinal neoplasms, Bmi1- or Lgr5-positive cells represent a population of cancer stem cells, whereas Lgr5-positive cells also function as cells-of-origin for intestinal tumors.


Asunto(s)
Biomarcadores de Tumor/genética , Evolución Clonal , Neoplasias Intestinales/genética , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Células Cultivadas , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/citología , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
8.
Sci Rep ; 6: 39386, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-28004815

RESUMEN

We recently reported that the polycomb complex protein Bmi1 is a marker for lingual epithelial stem cells (LESCs), which are involved in the long-term maintenance of lingual epithelial tissue in the physiological state. However, the precise role of LESCs in generating tongue tumors and Bmi1-positive cell lineage dynamics in tongue cancers are unclear. Here, using a mouse model of chemically (4-nitroquinoline-1-oxide: 4-NQO) induced tongue cancer and the multicolor lineage tracing method, we found that each unit of the tumor was generated by a single cell and that the assembly of such cells formed a polyclonal tumor. Although many Bmi1-positive cells within the tongue cancer specimens failed to proliferate, some proliferated continuously and supplied tumor cells to the surrounding area. This process eventually led to the formation of areas derived from single cells after 1-3 months, as determined using the multicolor lineage tracing method, indicating that such cells could serve as cancer stem cells. These results indicate that LESCs could serve as the origin for tongue cancer and that cancer stem cells are present in tongue tumors.


Asunto(s)
Linaje de la Célula/fisiología , Epitelio/metabolismo , Células Madre Neoplásicas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias de la Lengua/metabolismo , 4-Nitroquinolina-1-Óxido/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Lengua/metabolismo , Neoplasias de la Lengua/inducido químicamente
9.
Biochem Biophys Res Commun ; 466(3): 333-8, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26362184

RESUMEN

The skin is responsible for a variety of physiological functions and is critical for wound healing and repair. Therefore, the regenerative capacity of the skin is important. However, stem cells responsible for maintaining the acral epithelium had not previously been identified. In this study, we identified the specific stem cells in the acral epithelium that participate in the long-term maintenance of sweat glands, ducts, and interadnexal epidermis and that facilitate the regeneration of these structures following injury. Lgr6-positive cells and Bmi1-positive cells were found to function as long-term multipotent stem cells that maintained the entire eccrine unit and the interadnexal epidermis. However, while Lgr6-positive cells were rapidly cycled and constantly supplied differentiated cells, Bmi1-positive cells were slow to cycle and occasionally entered the cell cycle under physiological conditions. Upon irradiation-induced injury, Bmi1-positive cells rapidly proliferated and regenerated injured epithelial tissue. Therefore, Bmi1-positive stem cells served as reservoir stem cells. Lgr5-positive cells were rapidly cycled and maintained only sweat glands; therefore, we concluded that these cells functioned as lineage-restricted progenitors. Taken together, our data demonstrated the identification of stem cells that maintained the entire acral epithelium and supported the different roles of three cellular classes.


Asunto(s)
Epitelio/metabolismo , Complejo Represivo Polycomb 1/fisiología , Proteínas Proto-Oncogénicas/fisiología , Receptores Acoplados a Proteínas G/fisiología , Células Madre/citología , Glándulas Sudoríparas/fisiología , Animales , Peso Corporal , Linaje de la Célula , Proliferación Celular , Epidermis/metabolismo , Femenino , Regulación de la Expresión Génica , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/citología , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Piel/metabolismo
10.
Exp Hematol ; 43(10): 849-57, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26021490

RESUMEN

Familial platelet disorder with propensity to acute myeloid leukemia (FPD/AML) is an autosomal dominant disease associated with a germline mutation in the RUNX1 gene and is characterized by thrombocytopenia and an increased risk of developing myeloid malignancies. We generated induced pluripotent stem cells (iPSCs) from dermal fibroblasts of a patient with FPD/AML possessing a nonsense mutation R174X in the RUNX1 gene. Consistent with the clinical characteristics of the disease, FPD iPSC-derived hematopoietic progenitor cells were significantly impaired in undergoing megakaryocytic differentiation and subsequent maturation, as determined by colony-forming cell assay and surface marker analysis. Notably, when we corrected the RUNX1 mutation using transcription activator-like effector nucleases in conjunction with a donor plasmid containing normal RUNX1 cDNA sequences, megakaryopoiesis and subsequent maturation were restored in FPD iPSC-derived hematopoietic cells. These findings clearly indicate that the RUNX1 mutation is robustly associated with thrombocytopenia in patients with FPD/AML, and transcription activator-like effector nuclease-mediated gene correction in iPSCs generated from patient-derived cells could provide a promising clinical application for treatment of the disease.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados , Trastornos de las Plaquetas Sanguíneas , Codón sin Sentido , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Terapia Genética/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mieloide Aguda , Trombopoyesis/genética , Adulto , Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/metabolismo , Trastornos de la Coagulación Sanguínea Heredados/patología , Trastornos de la Coagulación Sanguínea Heredados/terapia , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Trastornos de las Plaquetas Sanguíneas/patología , Trastornos de las Plaquetas Sanguíneas/terapia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN Complementario/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia
11.
Cancer Sci ; 106(5): 489-96, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25707772

RESUMEN

In recent years, several molecularly targeted therapies have been developed as part of lung cancer treatment; they have produced dramatically good results. However, among the many oncogenes that have been identified to be involved in the development of lung cancers, a number of oncogenes are not covered by these advanced therapies. For the treatment of lung cancers, which is a group of heterogeneous diseases, persistent effort in developing individual therapies based on the respective causal genes is important. In addition, for the development of a novel therapy, identification of the lung epithelial stem cells and the origin cells of lung cancer, and understanding about candidate cancer stem cells in lung cancer tissues, their intracellular signaling pathways, and the mechanism of dysregulation of the pathways in cancer cells are extremely important. However, the development of drug resistance by cancer cells, despite the use of molecularly targeted drugs for the causal genes, thus obstructing treatment, is a well-known phenomenon. In this article, we discuss major causal genes of lung cancers and intracellular signaling pathways involving those genes, and review studies on origin and stem cells of lung cancers, as well as the possibility of developing molecularly targeted therapies based on these studies.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Resistencia a Antineoplásicos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , MicroARNs/metabolismo , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Vía de Señalización Wnt
12.
Sci Rep ; 4: 6175, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25146451

RESUMEN

Asingle cells in undifferentiated spermatogonia are considered to be the most primitive forms of germ stem cells (GSCs). Although GFRα1 is thought to be a marker of Asingle cells, we found that Bmi1(High) is more specific than GFRα1 for Asingle cells. Bmi1(High) expression in Asingle cells is correlated with seminiferous stages, and its expression was followed by the proliferative stage of Asingle GSCs. In contrast, GFRα1 expression was seminiferous stage-independent. Fate analyses of EdU-positive Bmi1(High)-positive cell-derived Asingle cells revealed that these cells self-renewed or generated transient amplifying Apaired cells. Bmi1(High)-positive cells were resistant to irradiation-induced injury, after which they regenerated. Elimination of Bmi1(High)-positive cells from seminiferous tubules resulted in the appearance of tubules with seminiferous stage mismatches. Thus, in this study, we found that Bmi1(High) is a seminiferous stage-dependent marker for long-term GSCs and that Bmi1(High)-positive cells play important roles in maintaining GSCs and in regenerating spermatogenic progenitors after injury.


Asunto(s)
Expresión Génica , Células Germinativas/metabolismo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Células Madre/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Eliminación de Gen , Células Germinativas/citología , Células Germinativas/efectos de la radiación , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Inmunofenotipificación , Masculino , Ratones , Ratones Transgénicos , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Regeneración , Túbulos Seminíferos/citología , Espermatogonias/citología , Espermatogonias/metabolismo , Espermatogonias/efectos de la radiación , Células Madre/citología , Células Madre/efectos de la radiación
13.
Exp Hematol ; 42(9): 816-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24859480

RESUMEN

Induced pluripotent stem cells (iPS) derived from disease cells are expected to provide a new experimental material, especially for diseases from which samples are difficult to obtain. In this study, we generated iPS from samples from patients with primary and secondary myelofibrosis. The primary myelofibrosis cells had chromosome 13q deletions, and the secondary myelofibrosis (SMF) cells had JAK2V617F mutations. The myelofibrosis patient cell-derived iPS (MF-iPS) were confirmed as possessing these parental disease-specific genomic markers. The capacity to form three germ layers was confirmed by teratoma assay. By co-culture with specific feeder cells and cytokines, MF-iPS can re-differentiate into blood progenitor cells and finally into megakaryocytes. We found that mRNA levels of interleukin-8, one of the candidate cytokines related to the pathogenesis of myelofibrosis, was elevated predominantly in megakaryocytes derived from MF-iPS. Because megakaryocytes from myelofibrosis clones are considered to produce critical mediators to proliferate fibroblasts in the bone marrow and iPS can provide differentiated cells abundantly, the disease-specific iPS we established should be a good research tool for this intractable disease.


Asunto(s)
Deleción Cromosómica , Trastornos de los Cromosomas , Células Madre Pluripotentes Inducidas , Janus Quinasa 2 , Mutación Missense , Mielofibrosis Primaria , Adulto , Sustitución de Aminoácidos , Células Cultivadas , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 13/metabolismo , Técnicas de Cocultivo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Interleucina-8/genética , Interleucina-8/metabolismo , Janus Quinasa 2/genética , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Persona de Mediana Edad , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Teratoma
14.
Blood ; 123(16): 2540-9, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24574458

RESUMEN

Just as normal stem cells require niche cells for survival, leukemia-initiating cells (LICs) may also require niche cells for their maintenance. Chronic myeloid leukemia (CML) is caused by the activity of BCR-ABL, a constitutively active tyrosine kinase. CML therapy with tyrosine kinase inhibitors is highly effective; however, due to the persistence of residual LICs, it is not curative. Several factors are known to support CML LICs, but purification of LICs and a thorough understanding of their niche signals have not yet been achieved. Using a CML-like mouse model of myeloproliferative disease, we demonstrate that CML LICs can be divided into CD25(+)FcεRIα(-) Lineage marker (Lin)(-) Sca-1(+)c-Kit(+) (F(-)LSK) cells and CD25(-)F(-)LSK cells. The CD25(+)F(-)LSK cells had multilineage differentiation capacity, with a preference toward cytokine-producing mast cell commitment. Although cells interconverted between CD25(-)F(-)LSK and CD25(+)F(-)LSK status, the CD25(+)F(-)LSK cells exhibited higher LIC capacity. Our findings suggest that interleukin-2 derived from the microenvironment and CD25 expressed on CML LICs constitute a novel signaling axis. The high levels of CD25 expression in the CD34(+)CD38(-) fraction of human CML cells indicate that CD25(+) LICs constitute an "LIC-derived niche" that could be preferentially targeted in therapy for CML.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-2/fisiología , Interleucina-2/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Animales , Proliferación Celular , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Transducción de Señal/fisiología , Células Th2/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
15.
J Clin Invest ; 124(2): 528-42, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24382349

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy that originates from leukemia-initiating cells (LICs). The identification of common mechanisms underlying LIC development will be important in establishing broadly effective therapeutics for AML. Constitutive NF-κB pathway activation has been reported in different types of AML; however, the mechanism of NF-κB activation and its importance in leukemia progression are poorly understood. Here, we analyzed myeloid leukemia mouse models to assess NF-κB activity in AML LICs. We found that LICs, but not normal hematopoietic stem cells or non-LIC fractions within leukemia cells, exhibited constitutive NF-κB activity. This activity was maintained through autocrine TNF-α secretion, which formed an NF-κB/TNF-α positive feedback loop. LICs had increased levels of active proteasome machinery, which promoted the degradation of IκBα and further supported NF-κB activity. Pharmacological inhibition of the proteasome complex markedly suppressed leukemia progression in vivo. Conversely, enhanced activation of NF-κB signaling expanded LIC frequency within leukemia cell populations. We also demonstrated a strong correlation between NF-κB activity and TNF-α secretion in human AML samples. Our findings indicate that NF-κB/TNF-α signaling in LICs contributes to leukemia progression and provide a widely applicable approach for targeting LICs.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Transporte Activo de Núcleo Celular , Adulto , Anciano , Animales , Células de la Médula Ósea/metabolismo , Ácidos Borónicos/uso terapéutico , Bortezomib , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Leucemia/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Persona de Mediana Edad , Fenotipo , Complejo de la Endopetidasa Proteasomal/química , Pirazinas/uso terapéutico , Transducción de Señal , Factores de Tiempo
18.
Int J Hematol ; 98(2): 145-52, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23807288

RESUMEN

Induced pluripotent stem cells (iPSCs) can be generated from various types of cells with transduction of defined transcription factors. In addition to regenerative medicine, iPSCs have been used for the study of pathogenesis of inherited genetic diseases. Here, we presented the examples of the establishment of iPSCs from hematopoietic cells or fibroblasts from hematological disease patients. Hematopoietic cells would be a good donor source for establishing iPSCs owing to the high reprogramming efficiency. iPSCs can be generated not only from normal cells, but also from several types of tumor cells. However it is not so easy, because iPSCs from hematological malignancies have been established only from myeloproliferative neoplasms including chronic myelogenous leukemia (CML) and JAK2-V617F mutation-positive polycythemia vera (PV). iPSC technology has great potential to promote oncology research based on patient samples.


Asunto(s)
Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Janus Quinasa 2 , Leucemia Mielógena Crónica BCR-ABL Positiva , Células Madre Neoplásicas , Sustitución de Aminoácidos , Animales , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Mutación Missense , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología
19.
Blood ; 121(20): 4142-55, 2013 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-23547050

RESUMEN

Ecotropic viral integration site 1 (Evi1) is one of the master regulators in the development of acute myeloid leukemia (AML) and myelodysplastic syndrome. High expression of Evi1 is found in 10% of patients with AML and indicates a poor outcome. Several recent studies have indicated that Evi1 requires collaborative factors to induce AML. Therefore, the search for candidate factors that collaborate with Evi1 in leukemogenesis is one of the key issues in uncovering the mechanism of Evi1-related leukemia. Previously, we succeeded in making a mouse model of Evi1-related leukemia using a bone marrow transplantation (BMT) system. In the Evi1-induced leukemic cells, we identified frequent retroviral integrations near the CCAAT/enhancer-binding protein ß (C/EBPß) gene and overexpression of its protein. These findings imply that C/EBPß is a candidate gene that collaborates with Evi1 in leukemogenesis. Cotransduction of Evi1 and the shortest isoform of C/EBPß, liver inhibitory protein (LIP), induced AML with short latencies in a mouse BMT model. Overexpression of LIP alone also induced AML with longer latencies. However, excision of all 3 isoforms of C/EBPß (LAP*/LAP/LIP) did not inhibit the development of Evi1-induced leukemia. Therefore, isoform-specific intervention that targets LIP is required when we consider C/EBPß as a therapeutic target.


Asunto(s)
Trasplante de Médula Ósea , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide Aguda/genética , Proto-Oncogenes/fisiología , Factores de Transcripción/fisiología , Animales , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/patología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/patología , Proteína del Locus del Complejo MDS1 y EV11 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Proto-Oncogenes/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Blood ; 119(26): 6234-42, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22592606

RESUMEN

Induced pluripotent stem cells (iPSCs) can be generated by the expression of defined transcription factors not only from normal tissue, but also from malignant cells. Cancer-derived iPSCs are expected to provide a novel experimental opportunity to establish the disease model. We generated iPSCs from imatinib-sensitive chronic myelogenous leukemia (CML) patient samples. Remarkably, the CML-iPSCs were resistant to imatinib although they consistently expressed BCR-ABL oncoprotein. In CML-iPSCs, the phosphorylation of ERK1/2, AKT, and JNK, which are essential for the maintenance of both BCR-ABL (+) leukemia cells and iPSCs, were unchanged after imatinib treatment, whereas the phosphorylation of signal transducer and activator of transcription (STAT)5 and CRKL was significantly decreased. These results suggest that the signaling for iPSCs maintenance compensates for the inhibition of BCR-ABL. CML-iPSC-derived hematopoietic cells recovered the sensitivity to imatinib although CD34(+)38(-)90(+)45(+) immature cells were resistant to imatinib, which recapitulated the pathophysiologic feature of the initial CML. CML-iPSCs provide us with a novel platform to investigate CML pathogenesis on the basis of patient-derived samples.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Cultivo Primario de Células/métodos , Animales , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Análisis por Conglomerados , Técnicas de Cocultivo , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/fisiología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Análisis por Micromatrices , Modelos Teóricos , Morfolinas/farmacología , Nitrilos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...