Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 615(7950): 127-133, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36813966

RESUMO

Haematopoietic stem cells (HSCs) are a rare cell type that reconstitute the entire blood and immune systems after transplantation and can be used as a curative cell therapy for a variety of haematological diseases1,2. However, the low number of HSCs in the body makes both biological analyses and clinical application difficult, and the limited extent to which human HSCs can be expanded ex vivo remains a substantial barrier to the wider and safer therapeutic use of HSC transplantation3. Although various reagents have been tested in attempts to stimulate the expansion of human HSCs, cytokines have long been thought to be essential for supporting HSCs ex vivo4. Here we report the establishment of a culture system that allows the long-term ex vivo expansion of human HSCs, achieved through the complete replacement of exogenous cytokines and albumin with chemical agonists and a caprolactam-based polymer. A phosphoinositide 3-kinase activator, in combination with a thrombopoietin-receptor agonist and the pyrimidoindole derivative UM171, were sufficient to stimulate the expansion of umbilical cord blood HSCs that are capable of serial engraftment in xenotransplantation assays. Ex vivo HSC expansion was further supported by split-clone transplantation assays and single-cell RNA-sequencing analysis. Our chemically defined expansion culture system will help to advance clinical HSC therapies.


Assuntos
Técnicas de Cultura de Células , Proliferação de Células , Citocinas , Células-Tronco Hematopoéticas , Humanos , Proliferação de Células/efeitos dos fármacos , Células Clonais/citologia , Células Clonais/efeitos dos fármacos , Células Clonais/metabolismo , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Técnicas de Cultura de Células/métodos , Albuminas , Caprolactama , Polímeros , Receptores de Trombopoetina , Transplante Heterólogo , Análise da Expressão Gênica de Célula Única
2.
Nature ; 611(7935): 358-364, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36323784

RESUMO

The accumulation of senescent cells is a major cause of age-related inflammation and predisposes to a variety of age-related diseases1. However, little is known about the molecular basis underlying this accumulation and its potential as a target to ameliorate the ageing process. Here we show that senescent cells heterogeneously express the immune checkpoint protein programmed death-ligand 1 (PD-L1) and that PD-L1+ senescent cells accumulate with age in vivo. PD-L1- cells are sensitive to T cell surveillance, whereas PD-L1+ cells are resistant, even in the presence of senescence-associated secretory phenotypes (SASP). Single-cell analysis of p16+ cells in vivo revealed that PD-L1 expression correlated with higher levels of SASP. Consistent with this, administration of programmed cell death protein 1 (PD-1) antibody to naturally ageing mice or a mouse model with normal livers or induced nonalcoholic steatohepatitis reduces the total number of p16+ cells in vivo as well as the PD-L1+ population in an activated CD8+ T cell-dependent manner, ameliorating various ageing-related phenotypes. These results suggest that the heterogeneous expression of PD-L1 has an important role in the accumulation of senescent cells and inflammation associated with ageing, and the elimination of PD-L1+ senescent cells by immune checkpoint blockade may be a promising strategy for anti-ageing therapy.


Assuntos
Envelhecimento , Antígeno B7-H1 , Fenótipo , Receptor de Morte Celular Programada 1 , Animais , Camundongos , Envelhecimento/imunologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Inflamação/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Análise de Célula Única , Hepatopatia Gordurosa não Alcoólica , Fígado , Rejuvenescimento
3.
Cell ; 147(5): 1146-58, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22118468

RESUMO

Hematopoietic stem cells (HSCs) reside and self-renew in the bone marrow (BM) niche. Overall, the signaling that regulates stem cell dormancy in the HSC niche remains controversial. Here, we demonstrate that TGF-ß type II receptor-deficient HSCs show low-level Smad activation and impaired long-term repopulating activity, underlining the critical role of TGF-ß/Smad signaling in HSC maintenance. TGF-ß is produced as a latent form by a variety of cells, so we searched for those that express activator molecules for latent TGF-ß. Nonmyelinating Schwann cells in BM proved responsible for activation. These glial cells ensheathed autonomic nerves, expressed HSC niche factor genes, and were in contact with a substantial proportion of HSCs. Autonomic nerve denervation reduced the number of these active TGF-ß-producing cells and led to rapid loss of HSCs from BM. We propose that glial cells are components of a BM niche and maintain HSC hibernation by regulating activation of latent TGF-ß.


Assuntos
Medula Óssea/fisiologia , Células-Tronco Hematopoéticas/citologia , Células de Schwann/citologia , Fator de Crescimento Transformador beta3/metabolismo , Animais , Antígenos CD34/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/metabolismo , Células de Schwann/fisiologia , Simpatectomia
4.
Nature ; 571(7763): 117-121, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31142833

RESUMO

Multipotent self-renewing haematopoietic stem cells (HSCs) regenerate the adult blood system after transplantation1, which is a curative therapy for numerous diseases including immunodeficiencies and leukaemias2. Although substantial effort has been applied to identifying HSC maintenance factors through the characterization of the in vivo bone-marrow HSC microenvironment or niche3-5, stable ex vivo HSC expansion has previously been unattainable6,7. Here we describe the development of a defined, albumin-free culture system that supports the long-term ex vivo expansion of functional mouse HSCs. We used a systematic optimization approach, and found that high levels of thrombopoietin synergize with low levels of stem-cell factor and fibronectin to sustain HSC self-renewal. Serum albumin has long been recognized as a major source of biological contaminants in HSC cultures8; we identify polyvinyl alcohol as a functionally superior replacement for serum albumin that is compatible with good manufacturing practice. These conditions afford between 236- and 899-fold expansions of functional HSCs over 1 month, although analysis of clonally derived cultures suggests that there is considerable heterogeneity in the self-renewal capacity of HSCs ex vivo. Using this system, HSC cultures that are derived from only 50 cells robustly engraft in recipient mice without the normal requirement for toxic pre-conditioning (for example, radiation), which may be relevant for HSC transplantation in humans. These findings therefore have important implications for both basic HSC research and clinical haematology.


Assuntos
Técnicas de Cultura de Células/métodos , Autorrenovação Celular/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Clonais/citologia , Células Clonais/efeitos dos fármacos , Meios de Cultura/química , Meios de Cultura/farmacologia , Feminino , Fibronectinas/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Álcool de Polivinil/farmacologia , Albumina Sérica , Fator de Células-Tronco/farmacologia , Trombopoetina/farmacologia , Fatores de Tempo , Condicionamento Pré-Transplante
5.
Nature ; 571(7766): E12, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31289376

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

6.
Cancer Sci ; 115(3): 698-705, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38221718

RESUMO

Hematopoietic stem cells (HSCs) are a rare population of cells found in the bone marrow that play a critical role in lifelong hematopoiesis and the reconstitution of the hematopoietic system after hematopoietic stem cell transplantation. Hematopoietic stem cell transplantation remains the only curative treatment for patients with refractory hematologic disorders, and umbilical cord blood (CB) serves as an alternative stem cell source due to its several advantageous characteristics, including human leukocyte antigen flexibility and reduced donor burden. However, CB also has the disadvantage of containing a small number of cells, resulting in limited donor selection and a longer time for engraftment. Therefore, the development of techniques to expand HSCs ex vivo, particularly umbilical CB, is a goal in hematology. While various combinations of cytokines were once the mainstream approach, these protocols had limited expansion rates and did not lead to clinical application. However, in recent years, the development of a technique in which small molecules are added to cytokines has enabled the stable, long-term ex vivo expansion of human HSCs. Clinical trials of expanded umbilical CB using these techniques have been undertaken and have confirmed their efficacy and safety. In addition, we have successfully developed a recombinant-cytokine-free and albumin-free culture system for the long-term expansion of human HSCs. This approach could offer the potential for more selective expansion of human HSCs compared to previous protocols. This review discusses ex vivo culture protocols for expanding human HSCs and presents the results of clinical trials using these techniques, along with future perspectives.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Transplante de Células-Tronco Hematopoéticas/métodos , Citocinas , Diferenciação Celular , Hematopoese
7.
Genes Cells ; 28(10): 679-693, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37584256

RESUMO

The evolutionally conserved Cdc7 kinase plays crucial roles in initiation of DNA replication as well as in other chromosomal events. To examine the roles of Cdc7 in brain development, we have generated mice carrying Cdc7 knockout in neural stem cells by using Nestin-Cre. The Cdc7Fl/Fl NestinCre mice were born, but exhibited severe growth retardation and impaired postnatal brain development. These mice exhibited motor dysfunction within 9 days after birth and did not survive for more than 19 days. The cerebral cortical layer formation was impaired, although the cortical cell numbers were not altered in the mutant. In the cerebellum undergoing hypoplasia, granule cells (CGC) decreased in number in Cdc7Fl/F l NestinCre mice compared to the control at E15-18, suggesting that Cdc7 is required for DNA replication and cell proliferation of CGC at mid embryonic stage (before embryonic day 15). On the other hand, the Purkinje cell numbers were not altered but its layer formation was impaired in the mutant. These results indicate differential roles of Cdc7 in DNA replication/cell proliferation in brain. Furthermore, the defects of layer formation suggest a possibility that Cdc7 may play an additional role in cell migration during neural development.


Assuntos
Proteínas de Ciclo Celular , Proteínas Serina-Treonina Quinases , Animais , Camundongos , Proteínas de Ciclo Celular/metabolismo , Cerebelo/metabolismo , Replicação do DNA , Nestina/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
8.
EMBO Rep ; 23(10): e55502, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-35971894

RESUMO

Hematopoietic stem cells (HSCs) cultured outside the body are the fundamental component of a wide range of cellular and gene therapies. Recent efforts have achieved > 200-fold expansion of functional HSCs, but their molecular characterization has not been possible since the majority of cells are non-HSCs and single cell-initiated cultures have substantial clone-to-clone variability. Using the Fgd5 reporter mouse in combination with the EPCR surface marker, we report exclusive identification of HSCs from non-HSCs in expansion cultures. By directly linking single-clone functional transplantation data with single-clone gene expression profiling, we show that the molecular profile of expanded HSCs is similar to proliferating fetal HSCs and reveals a gene expression signature, including Esam, Prdm16, Fstl1, and Palld, that can identify functional HSCs from multiple cellular states. This "repopulation signature" (RepopSig) also enriches for HSCs in human datasets. Together, these findings demonstrate the power of integrating functional and molecular datasets to better derive meaningful gene signatures and opens the opportunity for a wide range of functional screening and molecular experiments previously not possible due to limited HSC numbers.


Assuntos
Proteínas Relacionadas à Folistatina , Animais , Células Cultivadas , Receptor de Proteína C Endotelial/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Fatores de Transcrição/metabolismo
9.
Pediatr Blood Cancer ; 71(3): e30812, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38078567

RESUMO

BACKGROUND: Glucocorticoids are associated with an increased risk of venous thrombosis. Glucocorticoid treatment increases coagulation factor and anticoagulant levels; however, its effect on hemostatic function remains unclear. This study aimed to investigate the changes in comprehensive coagulation profiles after glucocorticoid treatment in noninflammatory diseases to elucidate the direct contribution of glucocorticoids to hemostatic function. PROCEDURE: Patients diagnosed with primary immune thrombocytopenia requiring glucocorticoid treatment were prospectively enrolled in this study. Changes in coagulation factors and anticoagulants during glucocorticoid treatment and changes in thrombin generation potential were determined in the absence and presence of soluble thrombomodulin (sTM). RESULTS: Seven treatment cases (four for steroid pulse therapy and three for oral glucocorticoid therapy) in six patients with immune thrombocytopenia were examined. After glucocorticoid treatment, activated partial thromboplastin time significantly shortened, and activities of factor VIII, IX, XI, and XII significantly increased, except for von Willebrand factor antigen. Moreover, antithrombin and protein C (PC) activities significantly increased after glucocorticoid treatment. Two major parameters of thrombin generation potential, endogenous thrombin potential (ETP) and peak thrombin (Peak), significantly increased in the absence of sTM after glucocorticoid treatment. However, no significant increases in either parameter were observed in the presence of sTM. ETP-TM and Peak-TM ratios, which represent resistance to the anticoagulant effect of the PC pathway, significantly decreased after glucocorticoid treatment, suggesting that anticoagulant function via the PC pathway is elevated after glucocorticoid treatment. CONCLUSIONS: As glucocorticoids increase intrinsic coagulation factor and anticoagulant levels, hemostatic balance between pro- and anticoagulant functions is maintained.


Assuntos
Hemostáticos , Púrpura Trombocitopênica Idiopática , Humanos , Trombina/metabolismo , Anticoagulantes/uso terapêutico , Glucocorticoides/efeitos adversos , Fatores de Coagulação Sanguínea , Proteína C/metabolismo
10.
Nihon Shokakibyo Gakkai Zasshi ; 121(1): 63-70, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38220183

RESUMO

A woman in her 70s was admitted to our institution with complaints of right hypochondrium pain. Abdominal computed tomography revealed a 13-mm retroperitoneal tumor between the liver and right kidney. The tumor rapidly increased to 82mm within 2 months, a necrotic change was inside the tumor, and the inflammation spread to the surrounding diaphragm and the peritoneum. The patient underwent surgical resection including the affected diaphragm and the peritoneum. Histopathological examination revealed a myofibroblastic spindle-cell proliferation with prominent infiltration of inflammatory cells, such as the plasma cells, lymphocytes, neutrophils, and eosinophils, diagnosed as an inflammatory myofibroblastic tumor (IMT) based on positive smooth muscle actin staining. IMT arising from the retroperitoneum is a rare case in Japan;we report this case with literature review.


Assuntos
Granuloma de Células Plasmáticas , Neoplasias , Feminino , Humanos , Granuloma de Células Plasmáticas/patologia , Inflamação , Japão , Tomografia Computadorizada por Raios X , Idoso
11.
Mol Ther ; 30(2): 534-549, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34628050

RESUMO

We generated dual-antigen receptor (DR) T cells from induced pluripotent stem cells (iPSCs) to mitigate tumor antigen escape. These cells were engineered to express a chimeric antigen receptor (CAR) for the antigen cell surface latent membrane protein 1 (LMP1; LMP1-CAR) and a T cell receptor directed to cell surface latent membrane protein 2 (LMP2), in association with human leucocyte antigen A24, to treat therapy-refractory Epstein-Barr virus-associated lymphomas. We introduced LMP1-CAR into iPSCs derived from LMP2-specific cytotoxic T lymphocytes (CTLs) to generate rejuvenated CTLs (rejTs) active against LMP1 and LMP2, or DRrejTs. All DRrejT-treated mice survived >100 days. Furthermore, DRrejTs rejected follow-up inocula of lymphoma cells, demonstrating that DRrejTs persisted long-term. We also demonstrated that DRrejTs targeting CD19 and LMP2 antigens exhibited a robust tumor suppressive effect and conferred a clear survival advantage. Co-operative antitumor effect and in vivo persistence, with unlimited availability of DRrejT therapy, will provide powerful and sustainable T cell immunotherapy.


Assuntos
Infecções por Vírus Epstein-Barr , Células-Tronco Pluripotentes Induzidas , Linfoma , Receptores de Antígenos Quiméricos , Animais , Terapia Baseada em Transplante de Células e Tecidos , Herpesvirus Humano 4/genética , Imunoterapia Adotiva , Células-Tronco Pluripotentes Induzidas/metabolismo , Linfoma/genética , Linfoma/terapia , Camundongos , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T Citotóxicos , Proteínas da Matriz Viral/genética
12.
Clin Exp Nephrol ; 27(2): 161-170, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36336723

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is an ultra-rare and life-threatening disease. For decades, plasma therapy was used to manage patients with aHUS. Since eculizumab, a recombinant humanized anti-C5 monoclonal antibody, was approved for treatment of aHUS, it has been used to treat patients with aHUS. Here, we examined the effectiveness of eculizumab and plasma therapy, respectively in the treatment of pediatric patients with aHUS. METHODS: Data were collected from questionnaires sent to 75 institutions known to be treating thrombotic microangiopathy (TMA). RESULTS: A total of 24 patients were evaluable, in which no recurrence of TMA was reported at last observation. There were four therapy groups: two patients receiving supportive therapy, one receiving plasma therapy alone, 17 switching from plasma therapy to eculizumab (therapy switched), and four receiving eculizumab alone. Among 17 patients of therapy-switched group, only one patient achieved complete remission at the end of plasma therapy, 15 patients achieved complete remission after eculizumab initiation, and two patients reached end-stage renal disease. Adverse events were reported in nine cases; among these, meningococcal infection, anaphylaxis, and eculizumab-related infusion reaction were reported among those treated with eculizumab. CONCLUSION: This study provided substantial evidence from a Japanese population that the conversion from plasma therapy to eculizumab therapy should be considered in patients with aHUS who show an incomplete response to plasma therapy. In addition, although no new safety events were detected, careful attention should be paid to meningococcal infection, eculizumab-related infusion reactions and allergic reactions with administration of eculizumab.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Falência Renal Crônica , Microangiopatias Trombóticas , Humanos , Criança , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Anticorpos Monoclonais Humanizados/efeitos adversos , Microangiopatias Trombóticas/induzido quimicamente
13.
Aquaculture ; 562: 738781, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36091578

RESUMO

The COVID-19 pandemic and related policy responses have exacerbated global food and nutrition insecurity by disrupting supply chains and destroying livelihoods. Previous studies show that the impact has been particularly severe for poor populations with limited livelihood options, who already faced food insecurity before the pandemic struck. This paper describes how COVID-19-related policy responses have impacted low-income, subsistence-oriented fish farmers in Bhutan. Based on nationally representative data collected between November 2020 and January 2021, the paper documents the responses of 353 Bhutanese fish farmers to the altered operating conditions and market disruptions caused by COVID-19. Results indicate that these farmers' access to inputs such as fish seed have suffered substantial disruptions. However, on the output side of the supply chain, some farmers have made significant gains in terms of increased demand and higher fish prices in informal markets. Furthermore, the food security of most farmers has suffered minimal impact. Overall, Bhutanese fish farmers have proved to be robust to COVID-19-induced adverse outcomes relative to commercial aquaculture producers elsewhere. Small-scale, subsistence-oriented production of fish along with other crops has benefitted the Bhutanese fish farmers by shielding them from the negative economic outcomes associated with market shocks and by directly preserving their food security. Nevertheless, the fish farmers require critical support to access essential inputs and upscale or maintain production infrastructure, so that they can continue fish production during the COVID-19 pandemic and become more robust in the long run.

14.
Molecules ; 28(6)2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-36985787

RESUMO

The hard-to-culture slightly halophilic myxobacterium "Paraliomyxa miuraensis" SMH-27-4 produces antifungal cyclodepsipeptide miuraenamide A (1). Herein, the region (85.9 kbp) containing the biosynthetic gene cluster (BGC) coding the assembly of 1 was identified and heterologously expressed in Myxococcus xanthus. A biosynthetic pathway proposed using in silico analysis was verified through the gene disruption of the heterologous transformant. In addition to the core polyketide synthase (PKS) and nonribosomal peptide synthase (NRPS) genes, tyrosine halogenase and O-methyltransferase genes participated in the biosynthesis of 1 as their gene-disrupted mutants produced a new congener, debromomiuraenamide A (4), and a previously isolated congener, miuraenamide E (3), respectively. Multigene disruption provided a heterologous mutant that produced 1 with the highest yield among the prepared mutants. When fed on 3-bromo-L-tyrosine, this mutant produced more 1 in the yield of 1.21 mg/L, which was 20 times higher than that produced by the initially prepared heterologous transformant. Although this yield was comparable to that of the original producer SMH-27-4 (1 mg/L), the culture time was 4.5 times shorter than that of SMH-27-4, indicating a five-fold efficiency in productivity. The results indicate the great potential of the miuraenamide BGC for the future contribution to drug development through logical gene manipulation.


Assuntos
Depsipeptídeos , Myxococcales , Antibacterianos/farmacologia , Myxococcales/genética , Myxococcales/metabolismo , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo , Família Multigênica
15.
J Biol Chem ; 297(6): 101367, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34736895

RESUMO

Rif1 regulates DNA replication timing and double-strand break repair, and its depletion induces transcriptional bursting of two-cell (2C) zygote-specific genes in mouse ES cells. However, how Rif1 regulates zygotic transcription is unclear. We show here that Rif1 depletion promotes the formation of a unique Zscan4 enhancer structure harboring both histone H3 lysine 27 acetylation (H3K27ac) and moderate levels of silencing chromatin mark H3K9me3. Curiously, another enhancer mark H3K4me1 is missing, whereas DNA methylation is still maintained in the structure, which spreads across gene bodies and neighboring regions within the Zscan4 gene cluster. We also found by function analyses of Rif1 domains in ES cells that ectopic expression of Rif1 lacking N-terminal domain results in upregulation of 2C transcripts. This appears to be caused by dominant negative inhibition of endogenous Rif1 protein localization at the nuclear periphery through formation of hetero-oligomers between the N-terminally truncated and endogenous forms. Strikingly, in murine 2C embryos, most of Rif1-derived polypeptides are expressed as truncated forms in soluble nuclear or cytosolic fraction and are likely nonfunctional. Toward the morula stage, the full-length form of Rif1 gradually increased. Our results suggest that the absence of the functional full-length Rif1 due to its instability or alternative splicing and potential inactivation of Rif1 through dominant inhibition by N-terminally truncated Rif1 polypeptides may be involved in 2C-specific transcription program.


Assuntos
Replicação do DNA/fisiologia , Proteínas de Ligação a Telômeros/fisiologia , Ativação Transcricional/fisiologia , Zigoto/metabolismo , Acetilação , Animais , Cromatina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/genética , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Fatores de Transcrição/genética , Regulação para Cima
16.
Blood ; 136(14): 1670-1684, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492700

RESUMO

Additional sex combs-like 1 (ASXL1), an epigenetic modulator, is frequently mutated in myeloid neoplasms. Recent analyses of mutant ASXL1 conditional knockin (ASXL1-MT-KI) mice suggested that ASXL1-MT alone is insufficient for myeloid transformation. In our previous study, we used retrovirus-mediated insertional mutagenesis, which exhibited the susceptibility of ASXL1-MT-KI hematopoietic cells to transform into myeloid leukemia cells. In this screening, we identified the hematopoietically expressed homeobox (HHEX) gene as one of the common retrovirus integration sites. In this study, we investigated the potential cooperation between ASXL1-MT and HHEX in myeloid leukemogenesis. Expression of HHEX enhanced proliferation of ASXL1-MT-expressing HSPCs by inhibiting apoptosis and blocking differentiation, whereas it showed only modest effect in normal HSPCs. Moreover, ASXL1-MT and HHEX accelerated the development of RUNX1-ETO9a and FLT3-ITD leukemia. Conversely, HHEX depletion profoundly attenuated the colony-forming activity and leukemogenicity of ASXL1-MT-expressing leukemia cells. Mechanistically, we identified MYB and ETV5 as downstream targets for ASXL1-MT and HHEX by using transcriptome and chromatin immunoprecipitation-next-generation sequencing analyses. Moreover, we found that expression of ASXL1-MT enhanced the binding of HHEX to the promoter loci of MYB or ETV5 via reducing H2AK119ub. Depletion of MYB or ETV5 induced apoptosis or differentiation in ASXL1-MT-expressing leukemia cells, respectively. In addition, ectopic expression of MYB or ETV5 reversed the reduced colony-forming activity of HHEX-depleted ASXL1-MT-expressing leukemia cells. These findings indicate that the HHEX-MYB/ETV5 axis promotes myeloid transformation in ASXL1-mutated preleukemia cells.


Assuntos
Transformação Celular Neoplásica/genética , Predisposição Genética para Doença , Proteínas de Homeodomínio/genética , Mutação , Células Mieloides/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Apoptose/genética , Biomarcadores Tumorais , Biópsia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Ensaio de Unidades Formadoras de Colônias , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Estudos de Associação Genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imunofenotipagem , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/mortalidade , Leucemia Mieloide/patologia , Camundongos , Células Mieloides/patologia , Prognóstico , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo
17.
Nature ; 530(7589): 223-7, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26863982

RESUMO

Haematopoietic stem cells (HSCs) are arguably the most extensively characterized tissue stem cells. Since the identification of HSCs by prospective isolation, complex multi-parameter flow cytometric isolation of phenotypic subsets has facilitated studies on many aspects of HSC biology, including self-renewal, differentiation, ageing, niche, and diversity. Here we demonstrate by unbiased multi-step screening, identification of a single gene, homeobox B5 (Hoxb5, also known as Hox-2.1), with expression in the bone marrow that is limited to long-term (LT)-HSCs in mice. Using a mouse single-colour tri-mCherry reporter driven by endogenous Hoxb5 regulation, we show that only the Hoxb5(+) HSCs exhibit long-term reconstitution capacity after transplantation in primary transplant recipients and, notably, in secondary recipients. Only 7-35% of various previously defined immunophenotypic HSCs are LT-HSCs. Finally, by in situ imaging of mouse bone marrow, we show that >94% of LT-HSCs (Hoxb5(+)) are directly attached to VE-cadherin(+) cells, implicating the perivascular space as a near-homogenous location of LT-HSCs.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/metabolismo , Nicho de Células-Tronco , Animais , Antígenos CD/metabolismo , Biomarcadores/análise , Medula Óssea/metabolismo , Caderinas/metabolismo , Autorrenovação Celular , Regulação da Expressão Gênica , Genes Reporter/genética , Transplante de Células-Tronco Hematopoéticas , Proteínas de Homeodomínio/genética , Imunofenotipagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL
18.
Rinsho Ketsueki ; 63(10): 1422-1429, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-36351651

RESUMO

Hematopoietic stem cells (HSC) have self-renewal as well as multilineage differentiation capacity and maintain hematopoiesis throughout life. HSC transplantation (HSCT) is performed as a curative therapy for hematopoietic malignancies and nonmalignant hematopoietic disorders. Furthermore, bone marrow, mobilized peripheral blood, and cord blood are available sources for HSCT. HLA compatibility is the most critical factor for a successful HSCT. The HSC number in a graft is also invaluable for engraftment. Moreover, it is challenging to obtain an abundant number of HSC for patients with obesity, particularly, in cord blood. HSC ex vivo expansion is an appropriate solution for this problem. Extrinsic factors to expand and maintain HSCs, such as cytokines are identified from analysis of HSCs and their niche. Thus, HSC ex vivo expansion is improved by adding them in culture medium; however, it is still difficult for therapeutic applications. Recently, several small molecular compounds have been reported to facilitate ex vivo expansion of HSC. Clinical trials that transplant ex vivo expanded cord blood have been already expanded, and some trials demonstrate reduction of time to hematopoietic recovery. Thus, we anticipate that ex vivo expanded cord blood transplantation will be applied widely in the future.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Hematopoese , Proliferação de Células , Diferenciação Celular , Sangue Fetal
19.
Int Immunol ; 32(12): 785-798, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32840578

RESUMO

Toll-like receptors (TLRs) impact myeloid cell responsiveness to environmental cues such as pathogen components and metabolites. Although TLR protein expression in monocytes and tissue macrophages is thought to be optimized for microenvironments in each tissue, a comprehensive study has not been reported. We here examined protein expression of endogenous TLRs in tissue-resident myeloid cells. Neutrophils in peripheral blood, spleen, liver and lung expressed TLR2, TLR4 and TLR5 in all tissues. Ly6C+ MHC II‒ classical monocytes mature into Ly6C‒ MHC II+ monocyte-derived dendritic cells (moDCs) or Ly6C‒ MHC II‒ patrolling monocytes. These subsets were found in all the tissues studied. TLR2 and TLR4 were displayed on all of these subsets, regardless of location. In contrast, expression of endosomal TLRs did vary with tissues and subsets. moDCs expressed TLR9, but much less TLR7. In contrast, TLR7, not TLR3 or TLR9, was highly expressed in classical and patrolling monocytes. Tissue macrophages such as red pulp macrophages in the spleen, Kupffer cells in the liver, microglia in the brain, alveolar macrophages in the lung and adipose tissue macrophages all expressed TLR2, TLR4 and TLR3. TLR7 was also expressed in these tissue macrophages except Kupffer cells in the liver. TLR9 expression in tissue macrophages was much lower or hard to detect. These results suggest that expression of endosomal TLRs in myeloid cells is influenced by their differentiation status and tissue-specific microenvironments.


Assuntos
Endossomos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Receptores Toll-Like/imunologia , Animais , Células Cultivadas , Camundongos , Receptores Toll-Like/genética
20.
Cancer Sci ; 111(12): 4336-4347, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33037737

RESUMO

Monomer tubulin polymerize into microtubules, which are highly dynamic and play a critical role in mitosis. Therefore, microtubule dynamics are an important target for anticancer drugs. The inhibition of tubulin polymerization or depolymerization was previously targeted and exhibited efficacy against solid tumors. The novel small molecule PTC596 directly binds tubulin, inhibits microtubule polymerization, downregulates MCL-1, and induces p53-independent apoptosis in acute myeloid leukemia cells. We herein investigated the efficacy of PTC-028, a structural analog of PTC596, for myelodysplastic syndrome (MDS). PTC-028 suppressed growth and induced apoptosis in MDS cell lines. The efficacy of PTC028 in primary MDS samples was confirmed using cell proliferation assays. PTC-028 synergized with hypomethylating agents, such as decitabine and azacitidine, to inhibit growth and induce apoptosis in MDS cells. Mechanistically, a treatment with PTC-028 induced G2/M arrest followed by apoptotic cell death. We also assessed the efficacy of PTC-028 in a xenograft mouse model of MDS using the MDS cell line, MDS-L, and the AkaBLI bioluminescence imaging system, which is composed of AkaLumine-HCl and Akaluc. PTC-028 prolonged the survival of mice in xenograft models. The present results suggest a chemotherapeutic strategy for MDS through the disruption of microtubule dynamics in combination with DNA hypomethylating agents.


Assuntos
Benzimidazóis/farmacologia , Síndromes Mielodisplásicas/tratamento farmacológico , Pirazinas/farmacologia , Moduladores de Tubulina/farmacologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzimidazóis/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Decitabina/farmacologia , Fase G2/efeitos dos fármacos , Células HL-60 , Xenoenxertos , Humanos , Camundongos , Síndromes Mielodisplásicas/genética , Paclitaxel/farmacologia , Pirazinas/uso terapêutico , Análise de Sequência de RNA/métodos , Tubulina (Proteína)/efeitos dos fármacos , Moduladores de Tubulina/uso terapêutico , Vincristina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa