Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Sci ; 115(6): 2002-2011, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38498976

RESUMEN

Triplet regimen comprising proteasome inhibitors, immunomodulatory drugs, and dexamethasone (DEX) is a recommended induction/consolidation therapy for multiple myeloma (MM) patients eligible for transplant. In this Japanese phase II study conducted from 2017 to 2019, newly diagnosed MM patients aged 20-65 received four induction cycles with bortezomib (Bor), lenalidomide (Len), and DEX (VRD), followed by Bor and high-dose melphalan with autologous stem cell rescue. Subsequently, they underwent four consolidation cycles with carfilzomib, Len, and DEX (KRD), followed by Len maintenance until disease progression. A total of 141 patients were analyzed. In an intent-to-treat population, the complete or better response post induction was 19.9%, rising to 39.7%, 58.9%, and 62.4% after transplant, consolidation, and 1-year maintenance, respectively. With a median follow-up of 38 months, the 3-year progression-free survival (PFS) rate was 83.5% and the 3-year overall survival rate was 92.5%. Severe adverse events (≥grade 3) occurred in ~30% of patients; however, there was no treatment-related mortality. These findings clearly showed the tolerability and effectiveness of this protocol. Nevertheless, patients with high-risk cytogenetics showed a trend toward lower 3-year PFS than those without (77.8% vs. 89.4%, p = 0.051), and ultra-high-risk cytogenetics (≥2 high-risk cytogenetics) had an even worse prognosis, with 61.2% 3-year PFS. To overcome this situation, a more potent treatment strategy incorporating novel agents such as the CD38-antibody should be assessed in future studies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Bortezomib , Dexametasona , Trasplante de Células Madre Hematopoyéticas , Lenalidomida , Mieloma Múltiple , Trasplante Autólogo , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/terapia , Lenalidomida/administración & dosificación , Lenalidomida/uso terapéutico , Persona de Mediana Edad , Femenino , Masculino , Anciano , Adulto , Trasplante de Células Madre Hematopoyéticas/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Bortezomib/administración & dosificación , Bortezomib/uso terapéutico , Quimioterapia de Consolidación/métodos , Melfalán/administración & dosificación , Melfalán/uso terapéutico , Oligopéptidos/uso terapéutico , Oligopéptidos/administración & dosificación , Quimioterapia de Inducción/métodos , Supervivencia sin Progresión , Adulto Joven , Quimioterapia de Mantención/métodos
2.
Cancer Sci ; 114(8): 3247-3258, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37197765

RESUMEN

Metabolic alterations, especially in the mitochondria, play important roles in several kinds of cancers, including acute myeloid leukemia (AML). However, AML-specific molecular mechanisms that regulate mitochondrial dynamics remain elusive. Through the metabolite screening comparing CD34+ AML cells and healthy hematopoietic stem/progenitor cells, we identified enhanced lysophosphatidic acid (LPA) synthesis activity in AML. LPA is synthesized from glycerol-3-phosphate by glycerol-3-phosphate acyltransferases (GPATs), rate-limiting enzymes of the LPA synthesis pathway. Among the four isozymes of GPATs, glycerol-3-phosphate acyltransferases, mitochondrial (GPAM) was highly expressed in AML cells, and the inhibition of LPA synthesis by silencing GPAM or FSG67 (a GPAM-inhibitor) significantly impaired AML propagation through the induction of mitochondrial fission, resulting in the suppression of oxidative phosphorylation and the elevation of reactive oxygen species. Notably, inhibition of this metabolic synthesis pathway by FSG67 administration did not affect normal human hematopoiesis in vivo. Therefore, the GPAM-mediated LPA synthesis pathway from G3P represents a critical metabolic mechanism that specifically regulates mitochondrial dynamics in human AML, and GPAM is a promising potential therapeutic target.


Asunto(s)
Leucemia Mieloide Aguda , Dinámicas Mitocondriales , Humanos , Glicerol , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Aciltransferasas , Fosfatos
3.
Cancer Sci ; 114(7): 2895-2906, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36945114

RESUMEN

The cancer stem cell (CSC) theory features typically rare self-renewing subpopulations that reconstitute the heterogeneous tumor. Identification of molecules that characterize the features of CSCs is a key imperative for further understanding tumor heterogeneity and for the development of novel therapeutic strategies. However, the use of conventional markers of CSCs is still insufficient for the isolation of bona fide CSCs. We investigated organoids that are miniature forms of tumor tissues by reconstructing cellular diversity to identify specific markers to characterize CSCs in heterogeneous tumors. Here, we report that the receptor for hyaluronan-mediated motility (RHAMM) expresses in a subpopulation of CD44+ conventional human colorectal CSC fraction. Single-cell transcriptomics of organoids highlighted RHAMM-positive proliferative cells that revealed distinct characteristics among the various cell types. Prospectively isolated RHAMM+CD44+ cells from the human colorectal cancer tissues showed highly proliferative characteristics with a self-renewal ability in comparison with the other cancer cells. Furthermore, inhibition of RHAMM strongly suppressed organoid formation in vitro and inhibited tumor growth in vivo. Our findings suggest that RHAMM is a potential therapeutic target because it is a specific marker of the proliferative subpopulation within the conventional CSC fraction.


Asunto(s)
Neoplasias Colorrectales , Receptores de Hialuranos , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Colorrectales/patología , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral
4.
Rinsho Ketsueki ; 64(6): 547-552, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-37407480

RESUMEN

Acute myeloid leukemia (AML) is one of the most common hematologic malignancies derived from self-renewing and highly propagating leukemic stem cells (LSCs). We have previously identified T-cell immunoglobulin mucin-3 (TIM-3) as an AML LSC-specific surface molecule by comparing the gene expression profiles of LSCs and hematopoietic stem cells (HSCs). TIM-3 expression clearly discriminates LSCs from HSCs within the CD34+CD38- stem cell fraction. Furthermore, AML cells secrete galectin-9 (Gal-9, a TIM-3 ligand) in an autocrine manner, resulting in constitutive TIM-3 signaling, which maintains LSC self-renewal capacity through ß-catenin accumulation. In this study, we investigated the LSC-specific mechanisms of TIM-3 signaling. We found that TIM-3 signaling drove the canonical Wnt pathway, which was independent of Wnt ligands, to maintain cancer stemness in LSCs. Gal-9 ligation activated the cytoplasmic Src homology 2 (SH2) binding domain of TIM-3 to recruit hematopoietic cell kinase (HCK), a Src family kinase that is highly expressed in LSCs. HCK phosphorylated p120-catenin to promote the formation of the LDL receptor-related protein 6 (LRP6) signalosome, hijacking the canonical Wnt pathway. This TIM-3/HCK/p120-catenin axis was employed principally in immature LSCs compared to TIM-3-expressing exhausted T-cells.


Asunto(s)
Leucemia Mieloide Aguda , Vía de Señalización Wnt , Humanos , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , beta Catenina/metabolismo , Leucemia Mieloide Aguda/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Neoplásicas
5.
Blood ; 135(19): 1661-1672, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32206775

RESUMEN

In human-to-mouse xenogeneic transplantation, polymorphisms of signal-regulatory protein α (SIRPA) that decide their binding affinity for human CD47 are critical for engraftment efficiency of human cells. In this study, we generated a new C57BL/6.Rag2nullIl2rgnull (BRG) mouse line with Sirpahuman/human (BRGShuman) mice, in which mouse Sirpa was replaced by human SIRPA encompassing all 8 exons. Macrophages from C57BL/6 mice harboring Sirpahuman/human had a significantly stronger affinity for human CD47 than those harboring SirpaNOD/NOD and did not show detectable phagocytosis against human hematopoietic stem cells. In turn, Sirpahuman/human macrophages had a moderate affinity for mouse CD47, and BRGShuman mice did not exhibit the blood cytopenia that was seen in Sirpa-/- mice. In human to mouse xenograft experiments, BRGShuman mice showed significantly greater engraftment and maintenance of human hematopoiesis with a high level of myeloid reconstitution, as well as improved reconstitution in peripheral tissues, compared with BRG mice harboring SirpaNOD/NOD (BRGSNOD). BRGShuman mice also showed significantly enhanced engraftment and growth of acute myeloid leukemia and subcutaneously transplanted human colon cancer cells compared with BRGSNOD mice. BRGShuman mice should be a useful basic line for establishing a more authentic xenotransplantation model to study normal and malignant human stem cells.


Asunto(s)
Antígenos de Diferenciación/fisiología , Neoplasias del Colon/patología , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Fagocitosis , Receptores Inmunológicos/fisiología , Animales , Apoptosis , Proliferación Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Femenino , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Células Madre Neoplásicas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood ; 135(24): 2146-2158, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32219443

RESUMEN

Immunomodulatory drugs (IMiDs) are key agents for the treatment of multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the recruitment of neosubstrates to cereblon, a substrate receptor of the E3 ubiquitin ligase complex; therefore, identification of cell-specific neosubstrates is important to understand the effects of IMiDs. In clinical practice, IMiDs induce thrombocytopenia, which frequently results in the discontinuation of IMiD treatment. In the current study, we sought to identify the molecular mechanism underlying thrombocytopenia induced by IMiD treatment. We found that IMiDs strongly impaired proplatelet formation, a critical step in functional platelet production, through the inhibition of autocrine estradiol signaling in human megakaryocytes. Furthermore, we identified aromatase, an indispensable enzyme for estradiol biosynthesis, as a novel neosubstrate of cereblon. IMiDs promoted the recruitment of aromatase to cereblon, resulting in the degradation of aromatase in a proteasome-dependent manner. Finally, aromatase was significantly degraded in the bone marrow of patients with multiple myeloma who developed thrombocytopenia with IMiD treatment. These data suggest that aromatase is a neosubstrate of cereblon that is responsible for IMiD-induced thrombocytopenia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aromatasa/metabolismo , Factores Inmunológicos/efectos adversos , Trombocitopenia/inducido químicamente , Trombocitopenia/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Anciano , Anciano de 80 o más Años , Femenino , Células HEK293 , Humanos , Células K562 , Masculino , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Mielopoyesis/efectos de los fármacos , Mielopoyesis/fisiología , Especificidad por Sustrato , Trombocitopenia/patología
7.
Cancer Sci ; 112(9): 3419-3426, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34159709

RESUMEN

Acute myeloid leukemia (AML) is hierarchically organized by self-renewing leukemic stem cells (LSCs). LSCs originate from hematopoietic stem cells (HSCs) by acquiring multistep leukemogenic events. To specifically eradicate LSCs, while keeping normal HSCs intact, the discrimination of LSCs from HSCs is important. We have identified T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) as an LSC-specific surface molecule in human myeloid malignancies and demonstrated its essential function in maintaining the self-renewal ability of LSCs. TIM-3 has been intensively investigated as a "coinhibitory" or "immune checkpoint" molecule of T cells. However, little is known about its distinct function in T cells and myeloid malignancies. In this review, we discuss the structure of TIM-3 and its function in normal blood cells and LSCs, emphasizing the specific signaling pathways involved, as well as the therapeutic applications of TIM-3 molecules in human myeloid malignancies.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/química , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Animales , Humanos , Leucemia Mieloide Aguda/patología , Estructura Molecular , Linfocitos T/metabolismo
8.
Cancer Sci ; 111(7): 2361-2373, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32314454

RESUMEN

To elucidate dynamic changes in native BCR-ABL and alternatively spliced tyrosine kinase inhibitor (TKI)-resistant but function-dead BCR-ABLIns35bp variant, following commencement or discontinuation of TKI therapy, each transcript was serially quantified in patients with chronic myeloid leukemia (CML) by deep sequencing. Because both transcripts were amplified together using conventional PCR system for measuring International Scale (IS), deep sequencing method was used for quantifying such BCR-ABL variants. At the initial diagnosis, 7 of 9 patients presented a small fraction of cells possessing BCR-ABLIns35bp , accounting for 0.8% of the total IS BCR-ABL, corresponding to actual BCR-ABLIns35bp value of 1.1539% IS. TKI rapidly decreased native BCR-ABL but not BCR-ABLIns35bp , leading to the initial increase in the proportion of BCR-ABLIns35bp . Thereafter, both native BCR-ABL and BCR-ABLIns35bp gradually decreased in the course of TKI treatment, whereas small populations positive for TKI-resistant BCR-ABLIns35bp continued fluctuating at low levels, possibly underestimating the molecular response (MR). Following TKI discontinuation, sequencing analysis of 54 patients revealed a rapid relapse, apparently derived from native BCR-ABL+ clones. However, IS fluctuating at low levels around MR4.0 marked a predominant persistence of cells expressing function-dead BCR-ABLIns35bp , suggesting that TKI resumption was unnecessary. We clarified the possible mechanism underlying mis-splicing BCR-ABLIns35bp , occurring at the particular pseudo-splice site within intron8, which can be augmented by TKI treatment through inhibition of RNA polymerase II phosphorylation. No mutations were found in spliceosomal genes. Therefore, monitoring IS functional BCR-ABL extracting BCR-ABLIns35bp would lead us to a correct evaluation of MR status, thus determining the adequate therapeutic intervention.


Asunto(s)
Empalme Alternativo , Proteínas de Fusión bcr-abl/genética , Mutación , Inhibidores de Proteínas Quinasas/farmacología , ARN Polimerasa II/metabolismo , Adulto , Anciano , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Sitios Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Intrones , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Masculino , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/uso terapéutico , ARN Polimerasa II/antagonistas & inhibidores , Análisis de la Célula Individual
9.
Rinsho Ketsueki ; 61(9): 1138-1149, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-33162509

RESUMEN

Genomics and novel molecularly targeted drugs for treating acute myelogenous leukemia (AML) are developing rapidly. To optimize the allocation of patients to the best possible treatment, we have to expedite test results of cytogenetic and molecular analyses for target mutations such as CBF and FLT3, since gene mutations are specifically associated with patient prognosis and therefore inform medical decision making. However, novel agents cannot completely eradicate AML because of the emergence of resistance to these agents; therefore, at the moment it is still necessary to combine cytotoxic treatment with novel agents. Hence, it becomes vital to understand how to stratify AML patients and subsequently treat the right patients with the right combination of cytotoxic treatments and novel agents.


Asunto(s)
Leucemia Mieloide Aguda , Adulto , Antineoplásicos/uso terapéutico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación , Pronóstico , Tirosina Quinasa 3 Similar a fms/genética
10.
Blood ; 129(25): 3332-3343, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28336526

RESUMEN

The developmental pathway for human megakaryocytes remains unclear, and the definition of pure unipotent megakaryocyte progenitor is still controversial. Using single-cell transcriptome analysis, we have identified a cluster of cells within immature hematopoietic stem- and progenitor-cell populations that specifically expresses genes related to the megakaryocyte lineage. We used CD41 as a positive marker to identify these cells within the CD34+CD38+IL-3RαdimCD45RA- common myeloid progenitor (CMP) population. These cells lacked erythroid and granulocyte-macrophage potential but exhibited robust differentiation into the megakaryocyte lineage at a high frequency, both in vivo and in vitro. The efficiency and expansion potential of these cells exceeded those of conventional bipotent megakaryocyte/erythrocyte progenitors. Accordingly, the CD41+ CMP was defined as a unipotent megakaryocyte progenitor (MegP) that is likely to represent the major pathway for human megakaryopoiesis, independent of canonical megakaryocyte-erythroid lineage bifurcation. In the bone marrow of patients with essential thrombocythemia, the MegP population was significantly expanded in the context of a high burden of Janus kinase 2 mutations. Thus, the prospectively isolatable and functionally homogeneous human MegP will be useful for the elucidation of the mechanisms underlying normal and malignant human hematopoiesis.


Asunto(s)
Hematopoyesis , Células Progenitoras de Megacariocitos/citología , Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/citología , Adulto , Animales , Antígenos CD/análisis , Linaje de la Célula , Células Cultivadas , Humanos , Células Progenitoras de Megacariocitos/patología , Megacariocitos/metabolismo , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Glicoproteína IIb de Membrana Plaquetaria/análisis , Transcriptoma
11.
Biol Blood Marrow Transplant ; 24(12): 2540-2548, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30026111

RESUMEN

Human herpes virus-6 (HHV6)-associated myelitis and calcineurin inhibitor-induced pain syndrome (CIPS) are serious complications of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Because these 2 complications cause similar sensory nerve-related symptoms, such as paresthesia, pruritus, and severe pain occurring around the engraftment, it can be difficult to differentially diagnose the 2 conditions. We retrospectively analyzed 435 recipients to distinguish clinical symptoms of these 2 complications. Twenty-four patients (5.5%) developed HHV6-associated encephalitis/myelitis; of these, 11 (2.5%) presented only with myelitis-related symptoms (HHV6-associated myelitis), which was confirmed by the detection of HHV6 DNA, and 8 (1.8%) had CIPS, with undetected HHV6 DNA. All patients with HHV6-associated myelitis or CIPS exhibited similar sensory nerve-related symptoms. Diagnostic images did not provide definite evidence specific for each disease. Symptoms of all patients with CIPS improved after switching to another immunosuppressant. Overall survival rate at 2 years for patients with HHV6-associated encephalitis/myelitis was significantly lower than that of CIPS (13.1% versus 29.2%; P = .049) or that of patients without HHV6-associated encephalitis/myelitis or CIPS (42.4%; P = .036), whereas there was no significant difference among the latter 2 groups (P = .889). The development of HHV6-associated encephalitis/myelitis but not CIPS was significantly associated with poor prognosis. Thus, transplant physicians should be aware that sensory nerve-related symptoms indicate early manifestations that might be correlated with reactivation of HHV6 or CIPS. Therefore, identification of HHV6 DNA is crucial for making a differential diagnosis and immediately starting appropriate treatments for each complication.


Asunto(s)
Inhibidores de la Calcineurina/efectos adversos , Encefalitis Viral/etiología , Herpesvirus Humano 6/patogenicidad , Dolor/inducido químicamente , Trasplante de Células Madre/efectos adversos , Trasplante Homólogo/efectos adversos , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dolor/patología , Adulto Joven
12.
Rinsho Ketsueki ; 58(10): 1838-1843, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28978822

RESUMEN

Hematopoietic stem cells (HSCs) have the potential to self-renew and differentiate into multi-lineage mature hematopoietic cells; thus, these cells can maintain hematopoiesis. Human HSCs reside within the CD34+CD38- cell fractions. Similarly, in acute myelogenous leukemia (AML), a small number of leukemic cells, called leukemic stem cells (LSCs), can be enriched within the identical CD34+CD38- cell fractions. LSCs can self-renew and produce clonogenic leukemic cells, whereas non-LSCs lack the potential to self-renew or maintain leukemia; thus, AML is organized as a hierarchy that originated from LSCs. LSCs play a central role in the maintenance and progression of leukemia; therefore, these cells should be an ultimate target for the eradication of human AML. Previous studies have revealed specific molecular machineries essential for LSCs. Targeting LSC-specific molecules should be a good therapeutic approach to kill LSCs without affecting normal cells. In this review, we would like to introduce the recent progress in the LSC study.


Asunto(s)
Células Madre Hematopoyéticas , Leucemia , Células Madre Neoplásicas , Animales , Humanos , Leucemia/diagnóstico , Pronóstico
13.
Rinsho Ketsueki ; 58(10): 1884-1894, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28978829

RESUMEN

Despite recent progress in diagnosis and leukemogenesis based on genomic landscapes in acute myelogenous leukemia (AML), advances in AML treatment lag behind. Over the past four decades, combination chemotherapy with anthracycline and cytarabine remains the standard induction therapy. Subsequent post-remission consolidation therapy stratifies patients into favorable-risk, intermediate-risk, and unfavorable-risk groups to assign post-remission therapies based on cytogenetic abnormalities and molecular mutations. Allogeneic stem-cell transplant decreases the risk of AML recurrence compared to standard chemotherapy, but it also raises the risk of serious complications. Recent large collections of matched genomic and clinical data may assist in selecting the best individualized therapy for each AML patient. Emerging evidence indicates that molecularly targeted therapies such as FLT3 and IDH inhibitors may be effective in distinct AML subtypes, providing further rationale for a personalized medicine approach. An umbrella trial, such as "BEAT AML Master Trial," designed to offer novel targeted therapy to AML patients based on their genetic characteristics, will be launching worldwide in the near future.


Asunto(s)
Leucemia Mieloide Aguda/terapia , Adulto , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Leucemia Mieloide Aguda/genética , Mutación , Factores de Riesgo
14.
Stem Cells ; 33(3): 976-87, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25446279

RESUMEN

Previous studies have predicted that reciprocal activation of GATA-1 and PU.1 regulates myelo-erythroid versus myelo-lymphoid lineage commitment in early hematopoiesis. Such PU.1-activating myelo-lymphoid progenitors exist within the lymphoid-primed multipotent progenitor (LMPP) population at the primitive Lineage(-) Sca-1(+) c-Kit(+) (LSK) stage. We here show that the counterpart of GATA-1-activating myelo-erythroid progenitor resides also at the LSK stage, expressing CD41 at a high level. Purified CD41(hi) LSK cells showed exceedingly strong and prolonged myelo-erythroid-restricted reconstitution, and primed myelo-erythroid gene expression with a more primitive molecular signature as compared to the original common myeloid progenitor (CMP). The CD41(hi) LSK cells more strongly contributed to emergent and malignant myelopoiesis than LMPPs, and produced the original CMP by downregulating Sca-1 and CD41, suggesting that they are the earliest CMPs. Thus, the hematopoietic developmental map should be revised by integrating the primary branchpoint comprised of the new, isolatable CD41(hi) CMP and the LMPP populations.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Leucemia Mieloide/patología , Células Progenitoras Mieloides/citología , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Células Cultivadas , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Progenitoras Mieloides/metabolismo
15.
Rinsho Ketsueki ; 57(4): 412-6, 2016 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-27169443

RESUMEN

Acute myeloid leukemia (AML) originates from self-renewing leukemic stem cells (LSCs), an ultimate therapeutic target for AML. We previously reported that the T-cell immunoglobulin mucin-3 (TIM-3) is expressed on the LCS surface in most types of AML. Since only the TIM-3(+), i.e. not the TIM-3(-), fraction of human AML cells can reconstitute human AML in immunodeficient mice, we hypothesized that the TIM-3 has an essential function in maintaining AML LSCs. Herein, we show that TIM-3 and its ligand, galectin-9 (Gal-9), constitute an autocrine loop critical for human AML LSC development. Serum Gal-9 was significantly elevated in primary AML patients and in mice xenografted with human AML. Neutralization of Gal-9 inhibited xenogeneic reconstitution of human AML, as well as Gal-9 ligation of TIM-3 co-activated NF-κB and ß-catenin signaling, suggesting that TIM-3 signaling is necessary for LSC self-renewal. Interestingly, identical changes were found to be involved in the progressive transformation of a variety of pre-leukemic disorders into myeloid leukemia. Thus, molecules constituting the TIM-3/Gal-9 autocrine loop are potential therapeutic targets applicable to most types of myeloid leukemia.


Asunto(s)
Comunicación Autocrina , Autorrenovación de las Células , Galectinas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Progresión de la Enfermedad , Galectinas/genética , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Leucemia Mieloide Aguda/patología
16.
Blood ; 121(5): 840-8, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23233661

RESUMEN

UNLABELLED: Transient marrow expansion of normal B-cell precursors, termed hematogones, is occasionally observed after hematopoietic stem cell transplantation (HSCT). To understand the clinical significance of this phenomenon, we enumerated hematogones in 108 consecutive patients who received allogeneic HSCT for the treatment of hematologic malignancies, including acute myelogenous leukemia, advanced myelodysplastic syndromes, acute lymphoblastic leukemia, and non-Hodgkin lymphoma. Hematogone quantitation was performed at the time of complete donor engraftment (median day 25 and 32 in patients who received bone marrow and cord blood cell transplants, respectively). Hematogones were polyclonal B cells, and their frequencies correlated positively with blood B-cell numbers, and inversely with donors' but not recipients' age, suggesting that hematogones reflect cell-intrinsic B-cell potential of donor cells. Interestingly, patients developing hematogones that comprised > 5% of bone marrow mononuclear cells constituted a group with significantly prolonged overall survival and relapse-free survival, irrespective of their primary disease or donor cell source. In addition, patients with > 5% hematogones developed severe acute graft-versus-host diseases less frequently, which may contribute toward their improved survival. We therefore conclude that the amount of hematogones at the time of engraftment may be a useful tool in predicting the prognosis of patients treated with allogeneic HSCT. KEY POINTS: Quantitation of hematogones at engraftment is useful to predict prognosis of patients treated with allogeneic stem cell transplantation.


Asunto(s)
Supervivencia de Injerto , Neoplasias Hematológicas , Trasplante de Células Madre Hematopoyéticas , Células Precursoras de Linfocitos B , Donante no Emparentado , Enfermedad Aguda , Adolescente , Adulto , Anciano , Trasplante de Médula Ósea , Trasplante de Células Madre de Sangre del Cordón Umbilical , Supervivencia sin Enfermedad , Femenino , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/terapia , Neoplasias Hematológicas/sangre , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/terapia , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Tasa de Supervivencia , Trasplante Homólogo
17.
Blood ; 121(8): 1316-25, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23293079

RESUMEN

Current mouse lines efficient for human cell xenotransplantation are backcrossed into NOD mice to introduce its multiple immunodeficient phenotypes. Our positional genetic study has located the NOD-specific polymorphic Sirpa as a molecule responsible for its high xenograft efficiency: it recognizes human CD47 and the resultant signaling may cause NOD macrophages not to engulf human grafts. In the present study, we established C57BL/6.Rag2(nullIl2rgnull) mice harboring NOD-Sirpa (BRGS). BRGS mice engrafted human hematopoiesis with an efficiency that was equal to or even better than that of the NOD.Rag1(nullIl2rgnull) strain, one of the best xenograft models. Consequently, BRGS mice are free from other NOD-related abnormalities; for example, they have normalized C5 function that enables the evaluation of complement-dependent cytotoxicity of antibodies against human grafts in the humanized mouse model. Our data show that efficient human cell engraftment found in NOD-based models is mounted solely by their polymorphic Sirpa. The simplified BRGS line should be very useful in future studies of human stem cell biology.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Receptores Inmunológicos/genética , Trasplante Heterólogo/inmunología , Trasplante Heterólogo/métodos , Animales , Anticuerpos Heterófilos/inmunología , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Linaje de la Célula/inmunología , Proteínas de Unión al ADN/genética , Femenino , Supervivencia de Injerto/inmunología , Hematopoyesis/fisiología , Humanos , Linfocitos/citología , Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Animales , Fagocitosis/inmunología , Polimorfismo Genético/inmunología , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal/inmunología
18.
Oncology ; 89 Suppl 1: 28-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26551150

RESUMEN

Acute myeloid leukemia (AML) originates from self-renewing leukemic stem cells (LSCs), an ultimate therapeutic target in AML. Eradication of LSCs should be a critical and efficient therapeutic approach for the cure of AML. T-cell immunoglobulin mucin-3 (TIM-3) is expressed in most types of AML LSCs, but not in normal hematopoietic stem cells (HSCs); therefore, TIM-3 would be one of the promising therapeutic targets to specifically kill AML LSCs, sparing normal HSCs. In xenograft models reconstituted with human AML LSCs or human normal HSCs, an anti-human TIM-3 mouse antibody with cytotoxic activities exerts a potent anti-leukemic effect by targeting AML LSCs but does not affect normal human hematopoiesis in vivo. Here, we would like to introduce the recent studies on TIM-3 in normal and malignant hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de la Membrana/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Animales , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Hematopoyesis , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Ratones , Receptores Virales/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nihon Rinsho ; 73(5): 811-5, 2015 May.
Artículo en Japonés | MEDLINE | ID: mdl-25985636

RESUMEN

Acute myelogenous leukemia (AML) originates from self-renewing leukemic stem cells (LSCs), an ultimate therapeutic target for AML. LSCs play a central role in the propagation of leukemia through their unique stem cell like properties, and LSCs share the many functional molecules with their normal counterpart hematopoietic stem cells (HSCs). For the establishment of LSCs-specific therapeutic approaches, it is quite important to understand the biological differences between LSCs and HSCs. Recent, studies have succeeded in clarifying these biological differences. In this review, I would like to introduce the biological significance of LSCs and discuss the molecular targeted therapy against LSCs.


Asunto(s)
Leucemia/tratamiento farmacológico , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Animales , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patología , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo
20.
Blood ; 120(19): 4058-67, 2012 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22990013

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is characterized by deregulated engulfment of hematopoietic stem cells (HSCs) by BM macrophages, which are activated presumably by systemic inflammatory hypercytokinemia. In the present study, we show that the pathogenesis of HLH involves impairment of the antiphagocytic system operated by an interaction between surface CD47 and signal regulatory protein α (SIRPA). In HLH patients, changes in expression levels and HLH-specific polymorphism of SIRPA were not found. In contrast, the expression of surface CD47 was down-regulated specifically in HSCs in association with exacerbation of HLH, but not in healthy subjects. The number of BM HSCs in HLH patients was reduced to approximately 20% of that of healthy controls and macrophages from normal donors aggressively engulfed HSCs purified from HLH patients, but not those from healthy controls in vitro. Furthermore, in response to inflammatory cytokines, normal HSCs, but not progenitors or mature blood cells, down-regulated CD47 sufficiently to be engulfed by macrophages. The expression of prophagocytic calreticulin was kept suppressed at the HSC stage in both HLH patients and healthy controls, even in the presence of inflammatory cytokines. These data suggest that the CD47-SIRPA antiphagocytic system plays a key role in the maintenance of HSCs and that its disruption by HSC-specific CD47 down-regulation might be critical for HLH development.


Asunto(s)
Antígeno CD47/metabolismo , Citofagocitosis/inmunología , Regulación hacia Abajo , Células Madre Hematopoyéticas/metabolismo , Linfohistiocitosis Hemofagocítica/etiología , ADP-Ribosil Ciclasa 1/metabolismo , Adolescente , Adulto , Anciano , Antígenos CD34/metabolismo , Antígenos de Diferenciación/genética , Antígeno CD47/genética , Línea Celular , Citocinas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Mediadores de Inflamación/farmacología , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Persona de Mediana Edad , Modelos Biológicos , Mutación , Interferencia de ARN , Receptores Inmunológicos/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA