Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
EMBO J ; 42(23): e113527, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37846891

RESUMEN

Emergency granulopoiesis is the enhanced and accelerated production of granulocytes that occurs during acute infection. The contribution of hematopoietic stem cells (HSCs) to this process was reported; however, how HSCs participate in emergency granulopoiesis remains elusive. Here, using a mouse model of emergency granulopoiesis we observe transcriptional changes in HSCs as early as 4 h after lipopolysaccharide (LPS) administration. We observe that the HSC identity is changed towards a myeloid-biased HSC and show that CD201 is enriched in lymphoid-biased HSCs. While CD201 expression under steady-state conditions reveals a lymphoid bias, under emergency granulopoiesis loss of CD201 marks the lymphoid-to-myeloid transcriptional switch. Mechanistically, we determine that lymphoid-biased CD201+ HSCs act as a first response during emergency granulopoiesis due to direct sensing of LPS by TLR4 and downstream activation of NF-κΒ signaling. The myeloid-biased CD201- HSC population responds indirectly during an acute infection by sensing G-CSF, increasing STAT3 phosphorylation, and upregulating LAP/LAP* C/EBPß isoforms. In conclusion, HSC subpopulations support early phases of emergency granulopoiesis due to their transcriptional rewiring from a lymphoid-biased to myeloid-biased population and thus establishing alternative paths to supply elevated numbers of granulocytes.


Asunto(s)
Células Madre Hematopoyéticas , Lipopolisacáridos , Lipopolisacáridos/metabolismo , Hematopoyesis , Granulocitos/metabolismo
2.
Cell Rep ; 42(3): 112165, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36862552

RESUMEN

Inflammatory stimuli cause a state of emergency myelopoiesis leading to neutrophil-like monocyte expansion. However, their function, the committed precursors, or growth factors remain elusive. In this study we find that Ym1+Ly6Chi monocytes, an immunoregulatory entity of neutrophil-like monocytes, arise from progenitors of neutrophil 1 (proNeu1). Granulocyte-colony stimulating factor (G-CSF) favors the production of neutrophil-like monocytes through previously unknown CD81+CX3CR1lo monocyte precursors. GFI1 promotes the differentiation of proNeu2 from proNeu1 at the cost of producing neutrophil-like monocytes. The human counterpart of neutrophil-like monocytes that also expands in response to G-CSF is found in CD14+CD16- monocyte fraction. The human neutrophil-like monocytes are discriminated from CD14+CD16- classical monocytes by CXCR1 expression and the capacity to suppress T cell proliferation. Collectively, our findings suggest that the aberrant expansion of neutrophil-like monocytes under inflammatory conditions is a process conserved between mouse and human, which may be beneficial for the resolution of inflammation.


Asunto(s)
Monocitos , Neutrófilos , Ratones , Animales , Humanos , Monocitos/fisiología , Mielopoyesis , Diferenciación Celular , Factor Estimulante de Colonias de Granulocitos
3.
J Immunol ; 209(3): 498-509, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35840161

RESUMEN

The mononuclear phagocyte system (MPS), composed of monocytes/macrophages and dendritic cells (DCs), plays a critical role at the interface of the innate and adaptive immune systems. However, the simplicity of MPS has been challenged recently by discoveries of novel cellular components. In the current study, we identified the CD135+ subset of monocytes as a novel class of APCs in mice. CD135+ monocytes were readily found in the bone marrow, spleen, and peripheral blood at steady state, and they expressed markers specific to DCs, including MHC class II and CD209a, along with markers for monocytes/macrophages. In addition, this subset phagocytosed bacteria and activated naive T lymphocytes, fulfilling the criteria for APCs. CD135+ monocytes were derived directly from macrophage DC progenitors, not from common monocyte progenitors or other monocytes, suggesting that these are distinct from conventional monocytes. These findings facilitate our understanding of the MPS network that regulates immune responses for host defense.


Asunto(s)
Células Dendríticas , Monocitos , Animales , Diferenciación Celular , Macrófagos , Ratones , Sistema Mononuclear Fagocítico
4.
Cancer Discov ; 11(12): 3142-3157, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34193438

RESUMEN

Natural killer (NK) cells and T cells are key effectors of antitumor immune responses and major targets of checkpoint inhibitors. In multiple cancer types, we find that the expression of Wnt signaling potentiator R-spondin genes (e.g., RSPO3) is associated with favorable prognosis and positively correlates with gene signatures of both NK cells and T cells. Although endothelial cells and cancer-associated fibroblasts comprise the R-spondin 3-producing cells, NK cells and T cells correspondingly express the R-spondin 3 receptor LGR6 within the tumor microenvironment (TME). Exogenous expression or intratumor injection of R-spondin 3 in tumors enhanced the infiltration and function of cytotoxic effector cells, which led to tumor regression. NK cells and CD8+ T cells independently and cooperatively contributed to R-spondin 3-induced control of distinct tumor types. The effect of R-spondin 3 was mediated in part through upregulation of MYC and ribosomal biogenesis. Importantly, R-spondin 3 expression enhanced tumor sensitivity to anti-PD-1 therapy, thereby highlighting new therapeutic avenues. SIGNIFICANCE: Our study identifies novel targets in enhancing antitumor immunity and sensitizing immune checkpoint inhibition, which provides a rationale for developing new immunotherapies against cancers. It also offers mechanistic insights on Wnt signaling-mediated modulation of anticancer immunity in the TME and implications for a putative R-spondin-LGR6 axis in regulating NK-cell biology. This article is highlighted in the In This Issue feature, p. 2945.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Células Endoteliales , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Microambiente Tumoral
5.
Am J Chin Med ; 49(2): 461-485, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33641653

RESUMEN

Traditional Chinese Medicine (TCM) is a practical medicine based on thousands of years of medical practice in China. Arsenic dispensing powder (ADP) has been used as a treatment for MDS patients with a superior efficacy on anemia at Xiyuan Hospital of China Academy of Chinese Medical Sciences. In this study, we retrospectively analyzed MDS patients that received ADP treatment in the past 9 years and confirmed that ADP improves patients' anemia and prolongs overall survival in intermediate-risk MDS patients. Then, we used the MDS transgenic mice model and cell line to explore the drug mechanism. In normal and MDS cells, ADP does not show cellular toxicity but promotes differentiation. In mouse MDS models, we observed that ADP showed significant efficacy on promoting erythropoiesis. In the BFU-E and CFU-E assays, ADP could promote erythropoiesis not only in normal clones but also in MDS clones. Mechanistically, we found that ADP could downregulate HIF1A in MDS clones through upregulation of VHL, P53 and MDM2, which is involved in two parallel pathways to downregulate HIF1A. We also confirmed that ADP upregulates GATA factors in normal clones. Thus, our clinical and experimental studies indicate that ADP is a promising drug to promote erythropoiesis in both MDS and normal clones with a superior outcome than current regular therapies. ADP promotes erythropoiesis in myelodysplastic syndromes via downregulation of HIF1A and upregulation of GATA factors.


Asunto(s)
Arsenicales/farmacología , Medicamentos Herbarios Chinos/farmacología , Eritropoyesis/efectos de los fármacos , Factores de Transcripción GATA/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Animales , Línea Celular , Regulación hacia Abajo , Humanos , Ratones , Polvos , Estudios Retrospectivos , Regulación hacia Arriba
6.
Blood Adv ; 4(14): 3343-3356, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32717031

RESUMEN

The transcription factor CCAAT enhancer-binding protein ß (C/EBPß) is required for stress-induced granulopoiesis at the level of hematopoietic stem/progenitor cells (HSPCs); however, its role and mechanisms of action in HSPCs are unknown. In this study, we assessed the regulation and functions of C/EBPß in HSPCs, especially under stress conditions. After 5-fluorouracil treatment or bone marrow transplantation, Cebpb-/- HSPCs exhibited impaired cell-cycle activation and myeloid differentiation at the early and late phases of regeneration, respectively, whereas at steady state, Cebpb deficiency did not affect HSPCs. C/EBPß was upregulated in response to hematopoietic stress, especially in CD150high long term-hematopoietic stem cells (LT-HSCs). Intracellular flow cytometric analysis that detected distinct domains of C/EBPß revealed that, among the 3 isoforms of C/EBPß, liver-enriched inhibitory protein (LIP) was upregulated in LT-HSCs prior to liver-enriched activating protein (LAP)/LAP* during regeneration. Early upregulation of LIP promoted cell-cycle entry of LT-HSCs by positively regulating Myc and expanded the HSPCs pool. Subsequent myeloid differentiation of amplified HSPCs was mediated by LAP/LAP*, which were upregulated at a later phase of regeneration. Collectively, our findings show that stress-induced sequential upregulation of C/EBPß isoforms is critical for fine-tuning the proliferation and differentiation of regenerating HSPCs.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Animales , Proteína beta Potenciadora de Unión a CCAAT , Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Ratones , Isoformas de Proteínas/genética
7.
Mol Cells ; 43(2): 145-152, 2020 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-31964134

RESUMEN

RUNX1 plays an important role in the regulation of normal hematopoiesis. RUNX1 mutations are frequently found and have been intensively studied in hematological malignancies. Germline mutations in RUNX1 cause familial platelet disorder with predisposition to acute myeloid leukemia (FPD/AML). Somatic mutations of RUNX1 are observed in various types of hematological malignancies, such as AML, acute lymphoblastic leukemia (ALL), myelodysplastic syndromes (MDS), myeloproliferative neoplasm (MPN), chronic myelomonocytic leukemia (CMML), and congenital bone marrow failure (CBMF). Here, we systematically review the clinical and molecular characteristics of RUNX1 mutations, the mechanisms of pathogenesis caused by RUNX1 mutations, and potential therapeutic strategies to target RUNX1-mutated cases of hematological malignancies.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Neoplasias Hematológicas/genética , Humanos , Mutación
8.
Cancer Sci ; 110(5): 1510-1517, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30844107

RESUMEN

Since the first identification of hypoxic cells in sections of carcinomas in the 1950s, hypoxia has been known as a central hallmark of cancer cells and their microenvironment. Indeed, hypoxia benefits cancer cells in their growth, survival, and metastasis. The historical discovery of hypoxia-inducible factor-1α (HIF1A) in the early 1990s had a great influence on the field as many phenomena in hypoxia could be explained by HIF1A. However, not all regions or types of tumors are necessarily hypoxic. Thus, it is difficult to explain whole cancer pathobiology by hypoxia, especially in the early stage of cancer. Upregulation of glucose metabolism in cancer cells has been well known. Oxygen-independent glycolysis is activated in cancer cells even in the normoxia condition, which is known as the Warburg effect. Accumulating evidence and recent advances in cancer metabolism research suggest that hypoxia-independent mechanisms for HIF signaling activation is a hallmark for cancer. There are various mechanisms that generate pseudohypoxic conditions, even in normoxia. Given the importance of HIF1A for cancer pathobiology, the pseudohypoxia concept could shed light on the longstanding mystery of the Warburg effect and accelerate better understanding of the diverse phenomena seen in a variety of cancers.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Hipoxia de la Célula , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Ácido Láctico/metabolismo , Neoplasias/genética , Estabilidad Proteica , Ácido Pirúvico/metabolismo , Transducción de Señal
9.
Blood Adv ; 3(3): 476-488, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755436

RESUMEN

Even in the era of ABL tyrosine kinase inhibitors, eradication of chronic myeloid leukemia (CML) stem cells is necessary for complete cure of the disease. Interferon-α (IFN-α) has long been used for the treatment of chronic-phase CML, but its mechanisms of action against CML stem cells remain unclear. We found that IFN-α upregulated CCAAT/enhancer binding protein ß (C/EBPß) in BCR-ABL-expressing mouse cells by activating STAT1 and STAT5, which were recruited to a newly identified 3' distal enhancer of Cebpb that contains tandemly aligned IFN-γ-activated site elements. Suppression or deletion of the IFN-γ-activated site elements abrogated IFN-α-dependent upregulation of C/EBPß. IFN-α induced differentiation and exhaustion of CML stem cells, both in vitro and in vivo, in a C/EBPß-dependent manner. In addition, IFN-α upregulated C/EBPß and induced exhaustion of lineage- CD34+ cells from CML patients. Collectively, these results clearly indicate that C/EBPß is a critical mediator of IFN-α-induced differentiation and exhaustion of CML stem cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Animales , Humanos , Ratones
10.
Cancer Discov ; 8(11): 1438-1457, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30139811

RESUMEN

Myelodysplastic syndromes (MDS) are heterogeneous hematopoietic disorders that are incurable with conventional therapy. Their incidence is increasing with global population aging. Although many genetic, epigenetic, splicing, and metabolic aberrations have been identified in patients with MDS, their clinical features are quite similar. Here, we show that hypoxia-independent activation of hypoxia-inducible factor 1α (HIF1A) signaling is both necessary and sufficient to induce dysplastic and cytopenic MDS phenotypes. The HIF1A transcriptional signature is generally activated in MDS patient bone marrow stem/progenitors. Major MDS-associated mutations (Dnmt3a, Tet2, Asxl1, Runx1, and Mll1) activate the HIF1A signature. Although inducible activation of HIF1A signaling in hematopoietic cells is sufficient to induce MDS phenotypes, both genetic and chemical inhibition of HIF1A signaling rescues MDS phenotypes in a mouse model of MDS. These findings reveal HIF1A as a central pathobiologic mediator of MDS and as an effective therapeutic target for a broad spectrum of patients with MDS.Significance: We showed that dysregulation of HIF1A signaling could generate the clinically relevant diversity of MDS phenotypes by functioning as a signaling funnel for MDS driver mutations. This could resolve the disconnection between genotypes and phenotypes and provide a new clue as to how a variety of driver mutations cause common MDS phenotypes. Cancer Discov; 8(11); 1438-57. ©2018 AACR. See related commentary by Chen and Steidl, p. 1355 This article is highlighted in the In This Issue feature, p. 1333.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/fisiopatología , Síndromes Mielodisplásicos/patología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metaboloma , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo
11.
Rinsho Ketsueki ; 59(6): 784-792, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29973460

RESUMEN

Tyrosine kinase inhibitors (TKIs) have markedly improved the prognosis of patients with chronic-phase chronic myeloid leukemia (CML). However, the development of a novel therapeutic strategy that can target and eradicate CML stem cells remains an important requirement to achieve a complete cure for CML because these stem cells cause relapse and drug resistance even in patients receiving TKI treatment. Interferon-α (IFNα) has long been used for the treatment of chronic-phase CML, and its efficacy is now being re-evaluated as a therapeutic option. Some clinical studies have demonstrated the additive efficacy of IFNα in CML patients treated with TKI. However, the mechanism of action is not fully understood. In this review, we introduce our recent findings on the effects of IFNα on CML stem cells via the transcription factor CCAAT/enhancer binding protein ß, which regulates stress-induced hematopoiesis.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Interferón-alfa/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Células Madre Neoplásicas/efectos de los fármacos , Resistencia a Antineoplásicos , Humanos
12.
Biochem Biophys Res Commun ; 496(2): 490-496, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29309790

RESUMEN

The emergence of new molecular targeting agents has improved the prognosis of patients with multiple myeloma (MM). However, MM remains incurable because MM stem cells are likely resistant to these agents. Thus, it is important to further investigate the biology of MM stem cells, which reside in the hypoxic bone marrow niche. In this study, we established and investigated the characteristics of hypoxia-adapted MM (HA-MM) cells, which could proliferate for more than six months under hypoxic conditions (1% O2). The G0 fraction of HA-MM cells was larger than that of parental MM cells under normoxic conditions (20% O2). HA-MM cells possess enhanced tumorigenicity in primary and secondary transplantation studies. HA-MM cells also exhibited increased mRNA levels of stem cell markers and an enhanced self-renewal ability, and thus demonstrated characteristics of MM stem cells. These cells overexpressed phosphorylated Smad2, and treatment with a transforming growth factor (TGF)-ß/Smad signaling inhibitor decreased their clonogenicity in a replating assay. In conclusion, MM cells adapted to long-exposure of hypoxia exhibit stem cell characters with TGF-ß/Smad pathway activation.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/genética , Proteína Smad2/genética , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/genética , Animales , Biomarcadores de Tumor/metabolismo , Hipoxia de la Célula , Línea Celular Transformada , Línea Celular Tumoral , Femenino , Humanos , Inmunofenotipificación , Ratones , Ratones Endogámicos NOD , Mieloma Múltiple/metabolismo , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Trasplante de Neoplasias , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Células Madre/patología , Análisis de Supervivencia , Factor de Crecimiento Transformador beta/metabolismo
13.
Haematologica ; 103(1): 40-50, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051279

RESUMEN

Activated erythropoietin (EPO) receptor (EPOR) signaling causes erythrocytosis. The important role of macrophages for the erythroid expansion and differentiation process has been reported, both in baseline and stress erythropoiesis. However, the significance of EPOR signaling for regulation of macrophages contributing to erythropoiesis has not been fully understood. Here we show that EPOR signaling activation quickly expands both erythrocytes and macrophages in vivo in mouse models of primary and secondary erythrocytosis. To mimic the chimeric condition and expansion of the disease clone in the polycythemia vera patients, we combined Cre-inducible Jak2V617F/+ allele with LysM-Cre allele which expresses in mature myeloid cells and some of the HSC/Ps (LysM-Cre;Jak2V617F/+ mice). We also generated inducible EPO-mediated secondary erythrocytosis models using Alb-Cre, Rosa26-loxP-stop-loxP-rtTA, and doxycycline inducible EPAS1-double point mutant (DPM) alleles (Alb-Cre;DPM mice). Both models developed a similar degree of erythrocytosis. Macrophages were also increased in both models without increase of major inflammatory cytokines and chemokines. EPO administration also quickly induced these macrophages in wild-type mice before observable erythrocytosis. These findings suggest that EPOR signaling activation could induce not only erythroid cell expansion, but also macrophages. Surprisingly, an in vivo genetic approach indicated that most of those macrophages do not express EPOR, but erythroid cells and macrophages contacted tightly with each other. Given the importance of the central macrophages as a niche for erythropoiesis, further elucidation of the EPOR signaling mediated-regulatory mechanisms underlying macrophage induction might reveal a potential therapeutic target for erythrocytosis.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/metabolismo , Eritroblastos/metabolismo , Macrófagos/metabolismo , Policitemia/etiología , Policitemia/metabolismo , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Animales , Biomarcadores , Recuento de Células , Subunidad beta Común de los Receptores de Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Eritroblastos/efectos de los fármacos , Eritropoyetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Orden Génico , Genes Reporteros , Vectores Genéticos , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Transgénicos , Modelos Biológicos , Fenotipo , Policitemia/patología , Receptores de Eritropoyetina/genética
14.
Blood ; 130(16): 1809-1818, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-28807982

RESUMEN

The transcription factor CCAAT/enhancer-binding protein ß (C/EBPß) is highly expressed in monocytes/macrophages. However, its roles in monopoiesis are largely unknown. Here, we investigated the roles of C/EBPß in monopoiesis. Further subdivision of monocytes revealed that Cebpb messenger RNA was highly upregulated in Ly6C- monocytes in bone marrow. Accordingly, the number of Ly6C- monocytes was significantly reduced in Cebpb-/- mice. Bone marrow chimera experiments and Mx1-Cre-mediated deletion of Cebpb revealed a cell-intrinsic and monocyte-specific requirement for C/EBPß in monopoiesis. In Cebpb-/- mice, turnover of Ly6C- monocytes was highly accelerated and apoptosis of Ly6C- monocytes was increased. Expression of Csf1r, which encodes a receptor for macrophage colony-stimulating factor, was significantly reduced in Ly6C- monocytes of Cebpb-/- mice. C/EBPß bound to positive regulatory elements of Csf1r and promoted its transcription. Collectively, these results indicate that C/EBPß is a critical factor for Ly6C- monocyte survival, at least in part through upregulation of Csf1r.


Asunto(s)
Apoptosis/genética , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Monocitos/fisiología , Animales , Antígenos Ly/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Células COS , Diferenciación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Chlorocebus aethiops , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Monocitos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología
16.
Int J Hematol ; 105(5): 587-597, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28044259

RESUMEN

The poor prognosis of adults with B cell precursor acute lymphoblastic leukemia (BCP-ALL) is attributed to leukemia cells that are protected by the bone marrow (BM) microenvironment. In the present study, we explored the pharmacological targeting of mesenchymal stromal/stem cells in BM (BM-MSCs) to eliminate chemoresistant BCP-ALL cells. Human BCP-ALL cells (NALM-6 cells) that adhered to human BM-MSCs (NALM-6/Ad) were highly resistant to multiple anti-cancer drugs, and exhibited pro-survival characteristics, such as an enhanced Akt/Bcl-2 pathway and increased populations in the G0 and G2/S/M cell cycle stages. Bortezomib, a proteasome inhibitor, interfered with adhesion between BM-MSCs and NALM-6 cells and up-regulated the matricellular protein SPARC (secreted protein acidic and rich in cysteine) in BM-MSCs, thereby reducing the NALM-6/Ad population. Inhibition of SPARC expression in BM-MSCs using a small interfering RNA enhanced adhesion of NALM-6 cells. Conversely, recombinant SPARC protein interfered with adhesion of NALM-6 cells. These results suggest that SPARC disrupts adhesion between BM-MSCs and NALM-6 cells. Co-treatment with bortezomib and doxorubicin prolonged the survival of BCP-ALL xenograft mice, with a significant reduction of leukemia cells in BM. Our findings demonstrate that bortezomib contributes to the elimination of BCP-ALL cells through disruption of their adhesion to BM-MSCs, and offer a novel therapeutic strategy for BCP-ALL through targeting of BM-MSCs.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/patología , Células de la Médula Ósea , Bortezomib/farmacología , Células Madre Mesenquimatosas , Terapia Molecular Dirigida , Osteonectina/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones SCID , Trasplante de Neoplasias , Osteonectina/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
17.
Biochem Biophys Res Commun ; 464(2): 654-8, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26168729

RESUMEN

The CCAAT/enhancer-binding protein ß (C/EBPß) transcription factor is required for granulopoiesis under stress conditions. However, little is known about its roles in steady state hematopoiesis. Here, we analyzed the peripheral blood and bone marrow of Cebpb(-/-) mice at steady state by flow cytometry and unexpectedly found that the number of peripheral blood monocytes was severely reduced, while the number of bone marrow monocytes was maintained. The ability of Cebpb(-/-) bone marrow cells to give rise to macrophages/monocytes in vitro was comparable to that of wild-type bone marrow cells. Apoptosis of monocytes was enhanced in the peripheral blood, but not in the bone marrow of Cebpb(-/-) mice. These results indicate that C/EBPß is required for the survival of monocytes in peripheral blood.


Asunto(s)
Apoptosis/fisiología , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Monocitos/citología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Recuento de Linfocitos , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL
18.
Int J Hematol ; 102(2): 218-29, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26121953

RESUMEN

The clinical application of mesenchymal stromal/stem cells (MSCs) has been extensively explored. In this study, we examined the availability of freshly donated umbilical cord blood (UCB) units that do not qualify for the Japanese banking system for transplantation because of their small volume as a source of MSCs. Forty-five UCB units were used. The median volume of each UCB unit and number of nucleated cells per unit were 40 mL and 5.39 × 10(8), respectively. MSCs were successfully isolated from 18 of 45 units (40 %). The MSC isolation rate was not affected by cell processing method or the interval between delivery and cell processing. The volume of the UCB unit and the mononuclear cell count were predictive factors of the MSC isolation rate. MSCs were effectively isolated by selecting UCB units with a volume of ≥54 mL and containing ≥1.28 × 10(8) mononuclear cells, yielding a MSC isolation rate of >70 %. UCB-derived MSCs were similar to bone marrow-derived MSCs in terms of their morphology, surface marker expression, and differentiation potential, apart from adipogenesis. Our data indicate that UCB units that are currently discarded due to inadequate volume should be reconsidered as a source of MSCs using the well-established UCB banking system.


Asunto(s)
Bancos de Muestras Biológicas , Separación Celular , Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo de Célula , Diferenciación Celular , Separación Celular/métodos , Femenino , Humanos , Inmunofenotipificación , Cariotipo , Masculino , Células Madre Mesenquimatosas/metabolismo , Sensibilidad y Especificidad
19.
Cancer Sci ; 106(7): 797-802, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25940801

RESUMEN

Steady-state hematopoiesis responds to extracellular stimuli to meet changing demands and also to pathologically altered intracellular signaling. Granulocyte production increases following infection or in response to cytokine stimulation, and activation of the CCAAT/enhancer-binding protein ß (C/EBPß) transcription factor is required for such stress-induced granulopoiesis, whereas C/EBPα plays a critical role in maintaining steady-state granulopoiesis. Different roles of these C/EBP transcription factors in different modes of hematopoiesis are evolutionally conserved from zebrafish to humans. In addition to reactions against infections, C/EBPß is responsible for cancer-driven myelopoiesis, which promotes cancer progression, at least in part, by abrogating the immune response in the cancer microenvironment. The BCR-ABL fusion protein activates emergency-specific pathway of granulopoiesis by upregulating C/EBPß. This in turn causes chronic phase chronic myeloid leukemia, which is characterized by myeloid expansion. The C/EBPß transcription factor also plays a role in other hematological malignancies of both myeloid and lymphoid lineage origin. Thus, elucidation of the upstream and downstream networks surrounding C/EBPß will lead to the development of novel therapeutic strategies for diseases mediated by non-steady-state hematopoiesis.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/fisiología , Hematopoyesis , Animales , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Neutropenia/congénito , Neutropenia/genética , Neutropenia/metabolismo
20.
Eur J Immunol ; 41(1): 67-75, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21182078

RESUMEN

Galectins comprise a family of animal lectins that differ in their affinity for ß-galactosides. Galectin-9 (Gal-9) is a tandem-repeat-type galectin that was recently shown to function as a ligand for T-cell immunoglobin domain and mucin domain-3 (Tim-3) expressed on terminally differentiated CD4(+) Th1 cells. Gal-9 modulates immune reactions, including the induction of apoptosis in Th1 cells. In this study, we investigated the effects of Gal-9 in murine models of acute GVH disease (aGVHD). First, we demonstrated that recombinant human Gal-9 inhibit MLR in a dose-dependent manner, involving both Ca(2+) influx and apoptosis in T cells. Next, we revealed that recombinant human Gal-9 significantly inhibit the progression of aGVHD in murine BM transplantation models. In conclusion, Gal-9 ameliorates aGVHD, possibly by inducing T-cell apoptosis, suggesting that gal-9 may be an attractive candidate for the treatment of aGVHD.


Asunto(s)
Apoptosis , Galectinas/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Enfermedad Aguda , Animales , Canales de Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...