Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
iScience ; 26(12): 108561, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38144459

RESUMO

Hematopoietic stem cell (HSC) surface markers improve the understanding of cell identity and function. Here, we report that human HSCs can be distinguished by their expression of the CEA Cell Adhesion Molecule 5 (CEACAM5, CD66e), which serves as a marker and a regulator of HSC function. CD66e+ cells exhibited a 5.5-fold enrichment for functional long term HSCs compared to CD66e- cells. CD66e+CD34+CD90+CD45RA- cells displayed robust multi-lineage repopulation and serial reconstitution ability in immunodeficient mice compared to CD66e-CD34+CD90+CD45RA-cells. CD66e expression also identified almost all repopulating HSCs within the CD34+CD90+CD45RA- population. Together, these results indicated that CEACAM5 is a marker that enriches functional human hematopoietic stem cells capable of long-term multi-lineage engraftment.

2.
Sci Rep ; 13(1): 14253, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37653079

RESUMO

Thrombocytopenia is a major and fatal complication in patients with acute myeloid leukemia (AML), which results from disrupted megakaryopoiesis by leukemic niche and blasts. Our previous research revealed that elevated interleukin-4 (IL-4) in AML bone marrow had adverse impact on multiple stages throughout megakaryopoiesis including hematopoietic stem cells (HSCs), but the specific mechanism remains unknown. In the present study, we performed single-cell transcriptome analysis and discovered activated oxidative stress pathway and apoptosis pathway in IL-4Rαhigh versus IL-4Rαlow HSCs. IL-4 stimulation in vitro led to apoptosis of HSCs and down-regulation of megakaryocyte-associated transcription factors. Functional assays displayed higher susceptibility of IL-4Rαhigh HSCs to tunicamycin and irradiation-induced apoptosis, demonstrating their vulnerability to endoplasmic reticulum (ER) stress injury. To clarify the downstream signaling of IL-4, we analyzed the transcriptomes of HSCs from AML bone marrow and found a remarkable down-regulation of the proteasome component Psmd13, whose expression was required for megakaryocytic-erythroid development but could be inhibited by IL-4 in vitro. We knocked down Psmd13 by shRNA in HSCs, and found their repopulating capacity and megakaryocytic differentiation were severely compromised, with increased apoptosis in vivo. In summary, our study uncovered a previous unrecognized regulatory role of IL-4-Psmd13 signaling in anti-stress and megakaryocytic differentiation capability of HSCs.


Assuntos
Células-Tronco Hematopoéticas , Interleucina-4 , Humanos , Interleucina-4/genética , Megacariócitos , Regulação para Baixo , Diferenciação Celular
3.
Sci Immunol ; 8(81): eabn6429, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36930730

RESUMO

Hematopoietic stem cell transplantation is an effective regenerative therapy for many malignant, inherited, or autoimmune diseases. However, our understanding of reconstituted hematopoiesis in transplant patients remains limited. Here, we uncover the reconstitution dynamics of human allogeneic hematopoietic stem and progenitor cells (HSPCs) at single-cell resolution after transplantation. Transplanted HSPCs underwent rapid and measurable changes during the first 30 days after transplantation, characterized by a strong proliferative response on the first day. Transcriptomic analysis of HSPCs enabled us to observe that immunoregulatory neutrophil progenitors expressing high levels of the S100A gene family were enriched in granulocyte colony-stimulating factor-mobilized peripheral blood stem cells. Transplant recipients who developed acute graft-versus-host disease (aGVHD) infused fewer S100Ahigh immunoregulatory neutrophil progenitors, immunophenotyped as Lin-CD34+CD66b+CD177+, than those who did not develop aGVHD. Therefore, our study provides insights into the regenerative process of transplanted HSPCs in human patients and identifies a potential criterion for identifying patients at high risk for developing aGVHD early after transplant.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Humanos , Fator Estimulador de Colônias de Granulócitos , Células-Tronco Hematopoéticas , Antígenos CD34/análise
4.
Dev Cell ; 57(24): 2745-2760.e6, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36493772

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) give rise to the blood system and maintain hematopoiesis throughout the human lifespan. Here, we report a transcriptional census of human bone-marrow-derived HSPCs from the neonate, infant, child, adult, and aging stages, showing two subpopulations of multipotent progenitors separated by CD52 expression. From birth to the adult stage, stem and multipotent progenitors shared similar transcriptional alterations, and erythroid potential was enhanced after the infant stage. By integrating transcriptome, chromatin accessibility, and functional data, we further showed that aging hematopoietic stem cells (HSCs) exhibited a bias toward megakaryocytic differentiation. Finally, in comparison with the HSCs from the cord blood, neonate bone-marrow-derived HSCs were more quiescent and had higher long-term regeneration capability and durable self-renewal. Taken together, this work provides an integral transcriptome landscape of HSPCs and identifies their dynamics in post-natal steady-state hemopoiesis, thereby helping explore hematopoiesis in development and diseases.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Criança , Humanos , Recém-Nascido , Diferenciação Celular , Células-Tronco Hematopoéticas/metabolismo , Lactente , Adulto , Idoso
5.
Haematologica ; 107(1): 154-166, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33299232

RESUMO

Hematopoietic stem cells (HSCs) are dominantly quiescent under homeostasis, which is a key mechanism of maintaining the HSC pool for life-long hematopoiesis. Dormant HSCs poise to be immediately activated on urgent conditions and can return to quiescence after regaining homeostasis. To date, the molecular networks of regulating the threshold of HSC dormancy, if exist, remain largely unknown. Here, we unveiled that deletion of Nupr1, a gene preferentially expressed in HSCs, activated the quiescence HSCs under homeostatic status, which conferred engraftment competitive advantage on HSCs without compromising their stemness and multi-lineage differentiation abilities in serial transplantation settings. Following an expansion protocol, the Nupr1-/- HSCs proliferate more robustly than their wild type counterparts in vitro. Nupr1 inhibits the expression of p53 and the rescue of which offsets the engraftment advantage. Our data unveil the de novo role of Nupr1 as an HSC quiescence-regulator, which provides insights into accelerating the engraftment efficacy of HSC transplantation by targeting the HSC quiescence-controlling network.


Assuntos
Proteínas de Ligação a DNA/genética , Células-Tronco Hematopoéticas , Proteínas de Neoplasias/genética , Proteína Supressora de Tumor p53 , Animais , Diferenciação Celular , Hematopoese/genética , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Proteína Supressora de Tumor p53/genética
6.
Natl Sci Rev ; 8(3): nwaa180, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34691592

RESUMO

High throughput single-cell RNA-seq has been successfully implemented to dissect the cellular and molecular features underlying hematopoiesis. However, an elaborate and comprehensive transcriptome reference of the whole blood system is lacking. Here, we profiled the transcriptomes of 7551 human blood cells representing 32 immunophenotypic cell types, including hematopoietic stem cells, progenitors and mature blood cells derived from 21 healthy donors. With high sequencing depth and coverage, we constructed a single-cell transcriptional atlas of blood cells (ABC) on the basis of both protein-coding genes and long noncoding RNAs (lncRNAs), and showed a high consistence between them. Notably, putative lncRNAs and transcription factors regulating hematopoietic cell differentiation were identified. While common transcription factor regulatory networks were activated in neutrophils and monocytes, lymphoid cells dramatically changed their regulatory networks during differentiation. Furthermore, we showed a subset of nucleated erythrocytes actively expressing immune signals, suggesting the existence of erythroid precursors with immune functions. Finally, a web portal offering transcriptome browsing and blood cell type prediction has been established. Thus, our work provides a transcriptional map of human blood cells at single-cell resolution, thereby offering a comprehensive reference for the exploration of physiological and pathological hematopoiesis.

7.
Blood ; 138(20): 1939-1952, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34388251

RESUMO

Adenosine-to-inosine RNA editing and the catalyzing enzyme adenosine deaminase are both essential for hematopoietic development and differentiation. However, the RNA editome during hematopoiesis and the underlying mechanisms are poorly defined. Here, we sorted 12 murine adult hematopoietic cell populations at different stages and identified 30 796 editing sites through RNA sequencing. The dynamic landscape of the RNA editome comprises stage- and group-specific and stable editing patterns, but undergoes significant changes during lineage commitment. Notably, we found that antizyme inhibitor 1 (Azin1) was highly edited in hematopoietic stem and progenitor cells (HSPCs). Azin1 editing results in an amino acid change to induce Azin1 protein (AZI) translocation to the nucleus, enhanced AZI binding affinity for DEAD box polypeptide 1 to alter the chromatin distribution of the latter, and altered expression of multiple hematopoietic regulators that ultimately promote HSPC differentiation. Our findings have delineated an essential role for Azin1 RNA editing in hematopoietic cells, and our data set is a valuable resource for studying RNA editing on a more general basis.


Assuntos
Proteínas de Transporte/genética , RNA Helicases DEAD-box/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Edição de RNA , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/metabolismo , Camundongos Endogâmicos C57BL , RNA/genética
8.
Blood ; 137(24): 3339-3350, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33881475

RESUMO

Total body irradiation (TBI) is commonly used in host conditioning regimens for human hematopoietic stem cell (HSC) transplantation to treat various hematological disorders. Exposure to TBI not only induces acute myelosuppression and immunosuppression, but also injures the various components of the HSC niche in recipients. Our previous study demonstrated that radiation-induced bystander effects (RIBE) of irradiated recipients decreased the long-term repopulating ability of transplanted mouse HSCs. However, RIBE on transplanted human HSCs have not been studied. Here, we report that RIBE impaired the long-term hematopoietic reconstitution of human HSCs as well as the colony-forming ability of human hematopoietic progenitor cells (HPCs). Our further analyses revealed that the RIBE-affected human hematopoietic cells showed enhanced DNA damage responses, cell-cycle arrest, and p53-dependent apoptosis, mainly because of oxidative stress. Moreover, multiple antioxidants could mitigate these bystander effects, though at different efficacies in vitro and in vivo. Taken together, these findings suggest that RIBE impair human HSCs and HPCs by oxidative DNA damage. This study provides definitive evidence for RIBE on transplanted human HSCs and further justifies the necessity of conducting clinical trials to evaluate different antioxidants to improve the efficacy of HSC transplantation for the patients with hematological or nonhematological disorders.


Assuntos
Efeito Espectador/efeitos dos fármacos , Dano ao DNA , Raios gama/efeitos adversos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Estresse Oxidativo/efeitos da radiação , Lesões Experimentais por Radiação/metabolismo , Animais , Feminino , Células-Tronco Hematopoéticas/patologia , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Lesões Experimentais por Radiação/patologia
9.
Oncol Rep ; 44(3): 1064-1074, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32705233

RESUMO

Long non­coding RNAs (lncRNAs) contribute to the tumorigeneses of numerous types of cancer, including glioma. The present study was designed to unveil a novel lncRNA functioning in glioma and explore the underlying mechanisms. lncRNA titin­antisense RNA1 (TTN­AS1), miR­27b­3p and Runt­related transcription factor 1 (RUNX1) expression in glioma tissues and cell lines was estimated by RT­qPCR. Si­TTN­AS1 was transfected into glioma cell lines (U251 and LN229), and CCK­8 assay, flow cytometry, wound healing and Transwell assays were applied to estimate the function of TTN­AS1 in glioma cells. miR­27b­3p inhibitor was used to explore the mechanisms. The results revealed that TTN­AS1 was highly expressed in glioma specimens and cell lines. Downregulation of TTN­AS1 inhibited the proliferation, migration and invasion of the glioma cells, as well as increased the rate of apoptosis. In vivo, the tumor growth was also inhibited by TTN­AS1 depletion in nude mice. Furthermore, we revealed that TTN­AS1 exerted oncogenic effects via sponging miR­27b­3p and thereby positively regulating RUNX1 expression. In conclusion, the present study supported that TTN­AS1 acts as an oncogene in glioma by targeting miR­27b­3p to release RUNX1. This finding may contribute to gene therapy of glioma.


Assuntos
Neoplasias Encefálicas/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Glioma/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Apoptose/genética , Encéfalo/patologia , Encéfalo/cirurgia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Glioma/cirurgia , Humanos , Camundongos , Oncogenes , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nat Cell Biol ; 22(6): 630-639, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32367048

RESUMO

How transplanted haematopoietic stem cells (HSCs) behave soon after they reside in a preconditioned host has not been studied due to technical limitations. Here, using single-cell RNA sequencing, we first obtained the transcriptome-based classifications of 28 haematopoietic cell types. We then applied them in conjunction with functional assays to track the dynamic changes of immunophenotypically purified HSCs in irradiated recipients within the first week after transplantation. Based on our transcriptional classifications, most homed HSCs in bone marrow and spleen became multipotent progenitors and, occasionally, some HSCs gave rise to megakaryocytic-erythroid or myeloid precursors. Parallel in vitro and in vivo functional experiments supported the paradigm of robust differentiation without substantial HSC expansion during the first week. Therefore, this study uncovers the previously inaccessible kinetics and fate choices of transplanted HSCs in myeloablated recipients at early stage, with implications for clinical applications of HSCs and other stem cells.


Assuntos
Diferenciação Celular , Células Precursoras Eritroides/citologia , Células-Tronco Hematopoéticas/citologia , Megacariócitos/citologia , Células Mieloides/citologia , Análise de Célula Única/métodos , Transcriptoma , Animais , Ciclo Celular , Linhagem da Célula , Células Precursoras Eritroides/metabolismo , Feminino , Células-Tronco Hematopoéticas/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo
11.
Leukemia ; 34(9): 2375-2383, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32094463

RESUMO

Bone marrow (BM) mesenchymal stem cells (MSCs) are critical components of the BM microenvironment and play an essential role in supporting hematopoiesis. Dysfunction of MSCs is associated with the impaired BM microenvironment that promotes leukemia development. However, whether and how restoration of the impaired BM microenvironment can inhibit leukemia development remain unknown. Using an established leukemia model and the RNA-Seq analysis, we discovered functional degeneration of MSCs during leukemia progression. Importantly, intra-BM instead of systemic transfusion of donor healthy MSCs restored the BM microenvironment, demonstrated by functional recovery of host MSCs, improvement of thrombopoiesis, and rebalance of myelopoiesis. Consequently, intra-BM MSC treatment reduced tumor burden and prolonged survival of the leukemia-bearing mice. Mechanistically, donor MSC treatment restored the function of host MSCs and reprogrammed host macrophages into arginase 1 positive phenotype with tissue-repair features. Transfusion of MSC-reprogrammed macrophages largely recapitulated the therapeutic effects of MSCs. Taken together, our study reveals that donor MSCs reprogram host macrophages to restore the BM microenvironment and inhibit leukemia development.


Assuntos
Leucemia/patologia , Macrófagos/patologia , Células-Tronco Mesenquimais/citologia , Microambiente Tumoral , Animais , Proliferação de Células , Reprogramação Celular , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL
13.
Bone Marrow Transplant ; 55(6): 1029-1040, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31804621

RESUMO

The bone marrow (BM) niche regulates multiple hematopoietic stem cell (HSC) processes. Clinical treatment for hematological malignancies by HSC transplantation often requires preconditioning via total body irradiation, which severely and irreversibly impairs the BM niche and HSC regeneration. Novel strategies are needed to enhance HSC regeneration in irradiated BM. We compared the effects of EGF, FGF2, and PDGFB on HSC regeneration using human mesenchymal stem cells (MSCs) that were transduced with these factors via lentiviral vectors. Among the above niche factors tested, MSCs transduced with PDGFB (PDGFB-MSCs) most significantly improved human HSC engraftment in immunodeficient mice. PDGFB-MSC-treated BM enhanced transplanted human HSC self-renewal in secondary transplantations more efficiently than GFP-transduced MSCs (GFP-MSCs). Gene set enrichment analysis showed increased antiapoptotic signaling in PDGFB-MSCs compared with GFP-MSCs. PDGFB-MSCs exhibited enhanced survival and expansion after transplantation, resulting in an enlarged humanized niche cell pool that provide a better humanized microenvironment to facilitate superior engraftment and proliferation of human hematopoietic cells. Our studies demonstrate the efficacy of PDGFB-MSCs in supporting human HSC engraftment.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Mesenquimais , Animais , Medula Óssea , Células-Tronco Hematopoéticas , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-sis
14.
Nat Commun ; 10(1): 5594, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31811153

RESUMO

Applying somatic cell reprogramming strategies in cancer cell biology is a powerful approach to analyze mechanisms of malignancy and develop new therapeutics. Here, we test whether leukemia cells can be reprogrammed in vivo using the canonical reprogramming transcription factors-Oct4, Sox2, Klf4, and c-Myc (termed as OSKM). Unexpectedly, we discover that OSKM can eradicate leukemia cells and dramatically improve survival of leukemia-bearing mice. By contrast, OSKM minimally impact normal hematopoietic cells. Using ATAC-seq, we find OSKM induce chromatin accessibility near genes encoding apoptotic regulators in leukemia cells. Moreover, this selective effect also involves downregulation of H3K9me3 as an early event. Dissection of the functional effects of OSKM shows that Klf4 and Sox2 play dominant roles compared to c-Myc and Oct4 in elimination of leukemia cells. These results reveal an intriguing paradigm by which OSKM-initiated reprogramming induction can be leveraged and diverged to develop novel anti-cancer strategies.


Assuntos
Apoptose/genética , Apoptose/fisiologia , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Leucemia/genética , Leucemia/metabolismo , Animais , Medula Óssea , Cromatina , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células THP-1
15.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 27(3): 950-957, 2019 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-31204960

RESUMO

OBJECTIVE: To observe the dynamic changes of hematopoietic reconstitution and multiple lineages differentiation at early phase after transplantation. METHODS: Whole bone marrow mononuclear cells (wBMMNC, 5×106) and enriched c-Kit+ hematopoietic stem/progenitor cells (HSPC, 3×105) from the BM of B6-Ly5.1 mice were transplanted into lethally irradiated B6-Ly5.2 mice, the frequencies and absolute numbers of donor-derived cells (including LKS- and LKS+) were detected by flow cytometry. The multiple lineages differentiation of donor-derived cells was also monitored by flow cytometry. The homing and early phase proliferations of donor-derived cells were observed by two-photon microscope. RESULTS: The donor-derived cells started to proliferation from 5-7 days after transplantation and reached the peak value at 2-3 weeks after wBMMNC transplantation. The donor-derived cells proliferated from 1-2 weeks and maintained until 4 weeks after c-kit+HSPC transplantation. At 1 week after transplantation, the donor-derived cells mainly differentiated into myeloid cells with a few lymphoid cells production (B cells) but the production of T cells was not observed at most in wBMMNC transplanted group, while myeloid cells occupied the majority of donor-derived cells at 2-4 weeks; donor-derived cells almost totally differentiated into myeloid cells at 1-3 weeks after transplantation in c-Kit+ transplanted group and donor-derived B cells appeared at 4 weeks. The absolute number of donor-derived LKS- and LKS+ cells in the BM of c-Kit+ transplanted group were much higher than that of wBMMNC group (P<0.001) at 2 weeks respectively. The clustering proliferation of cKit+ cells at 4-5 days after transplantation was observed by two photon microscope. CONCLUSION: The dynamical rate of proliferation and reconstitution of donor-derived cells are much earlier and quicker in c-Kit+ group than those of wBMMNC group. c-Kit+ cells mainly differentiate into myeloid cells within 1-3 weeks and the lymphoid cell differentiation starts at 4 weeks after transplantation. The immediate proliferation and differentiation of c-Kit+ cells within 1 week maybe due to the urgent needs of hematopoietic regeneration under the myeloablated hosts.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Animais , Transplante de Medula Óssea , Diferenciação Celular , Proliferação de Células , Camundongos , Camundongos Endogâmicos C57BL
16.
Haematologica ; 104(10): 1950-1961, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30792200

RESUMO

Normal hematopoiesis can be disrupted by the leukemic bone marrow microenvironment, which leads to cytopenia-associated symptoms including anemia, hemorrhage and infection. Thrombocytopenia is a major and sometimes fatal complication in patients with acute leukemia. However, the mechanisms underlying defective thrombopoiesis in leukemia have not been fully elucidated. In the steady state, platelets are continuously produced by megakaryocytes. Using an MLL-AF9-induced acute myeloid leukemia mouse model, we demonstrated a preserved number and proportion of megakaryocyte-primed hematopoietic stem cell subsets, but weakened megakaryocytic differentiation via both canonical and non-canonical routes. This primarily accounted for the dramatic reduction of megakaryocytic progenitors observed in acute myeloid leukemia bone marrow and a severe disruption of the maturation of megakaryocytes. Additionally, we discovered overproduction of interleukin-4 from bone marrow endothelial cells in acute myeloid leukemia and observed inhibitory effects of interleukin-4 throughout the process of megakaryopoiesis in vivo Furthermore, we observed that inhibition of interleukin-4 in combination with induction chemotherapy not only promoted recovery of platelet counts, but also prolonged the duration of remission in our acute myeloid leukemia mouse model. Our study elucidates a new link between interleukin-4 signaling and defective megakaryopoiesis in acute myeloid leukemia bone marrow, thereby offering a potential therapeutic target in acute myeloid leukemia.


Assuntos
Células da Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Interleucina-4/metabolismo , Leucemia Mieloide Aguda/metabolismo , Neoplasias Experimentais/metabolismo , Trombocitopenia/metabolismo , Animais , Células da Medula Óssea/patologia , Células Endoteliais/patologia , Interleucina-4/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Trombocitopenia/genética , Trombocitopenia/patologia
17.
Blood Adv ; 3(3): 419-431, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30733302

RESUMO

The cell of origin, defined as the normal cell in which the transformation event first occurs, is poorly identified in leukemia, despite its importance in understanding of leukemogenesis and improving leukemia therapy. Although hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were used for leukemia models, whether their self-renewal and differentiation potentials influence the initiation and development of leukemia is largely unknown. In this study, the self-renewal and differentiation potentials in 2 distinct types of HSCs (HSC1 [CD150+CD41-CD34-Lineage-Sca-1+c-Kit+ cells] and HSC2 [CD150-CD41-CD34-Lineage-Sca-1+c-Kit+ cells]) and 3 distinct types of HPCs (HPC1 [CD150+CD41+CD34-Lineage-Sca-1+c-Kit+ cells], HPC2 [CD150+CD41+CD34+Lineage-Sca-1+c-Kit+ cells], and HPC3 [CD150-CD41-CD34+Lineage-Sca-1+c-Kit+ cells]) were isolated from adult mouse bone marrow, and examined by competitive repopulation assay. Then, cells from each population were retrovirally transduced to initiate MLL-AF9 acute myelogenous leukemia (AML) and the intracellular domain of NOTCH-1 T-cell acute lymphoblastic leukemia (T-ALL). AML and T-ALL similarly developed from all HSC and HPC populations, suggesting multiple cellular origins of leukemia. New leukemic stem cells (LSCs) were also identified in these AML and T-ALL models. Notably, switching between immunophenotypical immature and mature LSCs was observed, suggesting that heterogeneous LSCs play a role in the expansion and maintenance of leukemia. Based on this mouse model study, we propose that acute leukemia arises from multiple cells of origin independent of the self-renewal and differentiation potentials in hematopoietic stem and progenitor cells and is amplified by LSC switchover.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/genética , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide Aguda/metabolismo , Camundongos , Células-Tronco/citologia
18.
Exp Hematol ; 60: 40-46.e2, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29307605

RESUMO

Impaired production of healthy hematopoietic cells from residual hematopoietic stem cells (HSCs) leads to high mortality in acute myeloid leukemia (AML). Previous studies have identified p21 and Egr3 as intrinsic factors responsible for the growth arrest and differentiation blockade of normal HSCs in leukemia; however, the related extrinsic factors remain unknown. In this study, we found that transforming growth factor ß (TGFß) signaling was upregulated in HSCs from bone marrow of mice with MLL-AF9-induced acute myeloid leukemia (AML) because of excessive production of TGFß1, especially from megakaryocytes, and overactivation of latent TGFß1 protein. We also found that SMAD3, a signal transducer of TGFß1, directly bound to Egr3 and upregulated its expression to arrest proliferation of HSCs. Our study provides evidence for targeting TGFß1 in AML to rectify normal hematopoiesis defects in clinical practice.


Assuntos
Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Megacariócitos/metabolismo , Proteínas de Neoplasias/biossíntese , Fator de Crescimento Transformador beta1/biossíntese , Animais , Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Megacariócitos/patologia , Camundongos , Proteínas de Neoplasias/genética , Fator de Crescimento Transformador beta1/genética
19.
Sheng Li Xue Bao ; 68(4): 423-34, 2016 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-27546503

RESUMO

As a classical type of tissue stem cells, hematopoietic stem cell (HSC) is the earliest discovered and has been widely applied in the clinic as a great successful example for stem cell therapy. Thus, HSC research represents a leading field in stem cell biology and regenerative medicine. Self-renewal, differentiation, quiescence, apoptosis and trafficking constitute major characteristics of functional HSCs. These characteristics also signify different dynamic states of HSC through physiological interactions with the microenvironment cues in vivo. This review covers our current knowledge on the physiological regulation of HSC and its underlying molecular mechanisms. It is our hope that this review will not only help our colleagues to understand how HSC is physiologically regulated but also serve as a good reference for the studies on stem cell and regenerative medicine in general.


Assuntos
Células-Tronco Hematopoéticas , Apoptose , Diferenciação Celular , Movimento Celular
20.
Exp Hematol ; 44(9): 874-885.e5, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27287689

RESUMO

The cyclin-dependent kinase inhibitor CDKN2C (p18(INK4c)) restrains self-renewal in hematopoietic stem cells (HSCs) and participates in the development and maturation of lymphoid cells. Deficiency in p18 predisposes mice and humans to hematopoietic lymphoid malignancies such as T-cell leukemia and multiple myeloma. However, the mechanism by which p18 regulates differentiation from HSCs to lymphoid cells is poorly understood. In this study, we found that a progenitor population characterized by its expression of surface markers, Lin(-) Sca-1(+) c-Kit(low) (LSK(low)), was markedly expanded in the bone marrow of p18 knock-out (p18(-/-)) mice. This novel population possessed lymphoid differentiation potential, but not myeloid differentiation potential, both in vitro and in vivo. Whereas LSK(low) cells and common lymphoid progenitors (CLPs) overlapped functionally in generating lymphoid cells, they were distinct cell populations, because they had different gene expression profiles. Unlike CLPs, LSK(low) cells did not express the interleukin-7 receptor. LSK(low) cells were derived from HSCs and were independent of the p18-deleted microenvironment. This cell population may represent a previously unappreciated transitional stage from HSCs to lymphoid progenitors that is strictly restricted by p18 under physiological conditions. Likewise, LSK(low) might serve as a new cellular target of lymphoid malignances in the absence of p18.


Assuntos
Inibidor de Quinase Dependente de Ciclina p18/metabolismo , Células Progenitoras Linfoides/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Animais , Biomarcadores , Transplante de Medula Óssea , Diferenciação Celular/genética , Linhagem da Célula/genética , Microambiente Celular , Análise por Conglomerados , Ensaio de Unidades Formadoras de Colônias , Inibidor de Quinase Dependente de Ciclina p18/genética , Perfilação da Expressão Gênica , Sobrevivência de Enxerto , Imunofenotipagem , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/metabolismo , Células Progenitoras Linfoides/citologia , Linfopoese , Camundongos , Camundongos Knockout , Modelos Biológicos , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA