7 results on '"HEMATOPOIETIC STEM"'
Search Results
2. Polycomb repressive complex 1.1 coordinates homeostatic and emergency myelopoiesis.
- Author
-
Nakajima-Takagi Y, Oshima M, Takano J, Koide S, Itokawa N, Uemura S, Yamashita M, Andoh S, Aoyama K, Isshiki Y, Shinoda D, Saraya A, Arai F, Yamaguchi K, Furukawa Y, Koseki H, Ikawa T, and Iwama A
- Subjects
- Animals, Mice, Histones, Cell Differentiation physiology, Hematopoietic Stem Cells metabolism, Polycomb Repressive Complex 1 metabolism, Myelopoiesis genetics
- Abstract
Polycomb repressive complex (PRC) 1 regulates stem cell fate by mediating mono-ubiquitination of histone H2A at lysine 119. While canonical PRC1 is critical for hematopoietic stem and progenitor cell (HSPC) maintenance, the role of non-canonical PRC1 in hematopoiesis remains elusive. PRC1.1, a non-canonical PRC1, consists of PCGF1, RING1B, KDM2B, and BCOR. We recently showed that PRC1.1 insufficiency induced by the loss of PCGF1 or BCOR causes myeloid-biased hematopoiesis and promotes transformation of hematopoietic cells in mice. Here we show that PRC1.1 serves as an epigenetic switch that coordinates homeostatic and emergency hematopoiesis. PRC1.1 maintains balanced output of steady-state hematopoiesis by restricting C/EBPα-dependent precocious myeloid differentiation of HSPCs and the HOXA9- and β-catenin-driven self-renewing network in myeloid progenitors. Upon regeneration, PRC1.1 is transiently inhibited to facilitate formation of granulocyte-macrophage progenitor (GMP) clusters, thereby promoting emergency myelopoiesis. Moreover, constitutive inactivation of PRC1.1 results in unchecked expansion of GMPs and eventual transformation. Collectively, our results define PRC1.1 as a novel critical regulator of emergency myelopoiesis, dysregulation of which leads to myeloid transformation., Competing Interests: YN, MO, JT, SK, NI, SU, MY, SA, KA, YI, DS, AS, FA, KY, YF, HK, TI, AI No competing interests declared, (© 2023, Nakajima-Takagi et al.)
- Published
- 2023
- Full Text
- View/download PDF
3. Uncovering perturbations in human hematopoiesis associated with healthy aging and myeloid malignancies at single-cell resolution.
- Author
-
Ainciburu M, Ezponda T, Berastegui N, Alfonso-Pierola A, Vilas-Zornoza A, San Martin-Uriz P, Alignani D, Lamo-Espinosa J, San-Julian M, Jiménez-Solas T, Lopez F, Muntion S, Sanchez-Guijo F, Molero A, Montoro J, Serrano G, Diaz-Mazkiaran A, Lasaga M, Gomez-Cabrero D, Diez-Campelo M, Valcarcel D, Hernaez M, Romero JP, and Prosper F
- Subjects
- Humans, Aged, Hematopoiesis, Cell Differentiation, Hematopoietic Stem Cells metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Homeodomain Proteins metabolism, Healthy Aging, Myelodysplastic Syndromes metabolism, Neoplasms pathology
- Abstract
Early hematopoiesis is a continuous process in which hematopoietic stem and progenitor cells (HSPCs) gradually differentiate toward specific lineages. Aging and myeloid malignant transformation are characterized by changes in the composition and regulation of HSPCs. In this study, we used single-cell RNA sequencing (scRNA-seq) to characterize an enriched population of human HSPCs obtained from young and elderly healthy individuals., Based on their transcriptional profile, we identified changes in the proportions of progenitor compartments during aging, and differences in their functionality, as evidenced by gene set enrichment analysis. Trajectory inference revealed that altered gene expression dynamics accompanied cell differentiation, which could explain aging-associated changes in hematopoiesis. Next, we focused on key regulators of transcription by constructing gene regulatory networks (GRNs) and detected regulons that were specifically active in elderly individuals. Using previous findings in healthy cells as a reference, we analyzed scRNA-seq data obtained from patients with myelodysplastic syndrome (MDS) and detected specific alterations of the expression dynamics of genes involved in erythroid differentiation in all patients with MDS such as TRIB2. In addition, the comparison between transcriptional programs and GRNs regulating normal HSPCs and MDS HSPCs allowed identification of regulons that were specifically active in MDS cases such as SMAD1, HOXA6, POU2F2, and RUNX1 suggesting a role of these transcription factors (TFs) in the pathogenesis of the disease., In summary, we demonstrate that the combination of single-cell technologies with computational analysis tools enable the study of a variety of cellular mechanisms involved in complex biological systems such as early hematopoiesis and can be used to dissect perturbed differentiation trajectories associated with perturbations such as aging and malignant transformation. Furthermore, the identification of abnormal regulatory mechanisms associated with myeloid malignancies could be exploited for personalized therapeutic approaches in individual patients., Competing Interests: MA, TE, NB, AA, AV, PS, DA, JL, MS, TJ, FL, SM, FS, AM, JM, GS, AD, ML, DG, MD, DV, MH, FP No competing interests declared, JR Employed by 10x Genomics since February 2021; this employment had no bearing on this work, (© 2023, Ainciburu, Ezponda et al.)
- Published
- 2023
- Full Text
- View/download PDF
4. Bone Marrow Transplantation Dynamics: When Progenitor Expansion Prevails.
- Author
-
Nakamura-Ishizu A, Ahmad SAI, and Suda T
- Subjects
- Cell Differentiation, Hematopoietic Stem Cells, Humans, Transcriptome, Bone Marrow Transplantation, Hematopoietic Stem Cell Transplantation
- Abstract
The transplantation of healthy hematopoietic stem cells (HSCs) contained in the bone marrow is a frontline treatment option for hematopoietic diseases. Detailed analysis of post-transplant HSC kinetics is crucial as the initial engraftment of HSCs influences prognosis. Dong et al. have explored the dynamic change in HSC cell fate upon bone marrow transplantation through the utilization of single-cell transcriptomic analysis., (Copyright © 2020. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
5. Macrophage Dysregulation and Impaired Skin Wound Healing in Diabetes.
- Author
-
Barman PK and Koh TJ
- Abstract
Monocytes (Mo) and macrophages (Mϕ) play important roles in normal skin wound healing, and dysregulation of wound Mo/Mϕ leads to impaired wound healing in diabetes. Although skin wound Mϕ originate both from tissue resident Mϕ and infiltrating bone marrow-derived Mo, the latter play dominant roles during the inflammatory phase of wound repair. Increased production of bone marrow Mo caused by alterations of hematopoietic stem and progenitor cell (HSPC) niche and epigenetic modifications of HSPCs likely contributes to the enhanced number of wound Mϕ in diabetes. In addition, an impaired transition of diabetic wound Mϕ from "pro-inflammatory" to "pro-healing" phenotypes driven by the local wound environment as well as intrinsic changes in bone marrow Mo is also thought to be partly responsible for impaired diabetic wound healing. The current brief review describes the origin, heterogeneity and function of wound Mϕ during normal skin wound healing followed by discussion of how dysregulated wound Mϕ numbers and phenotype are associated with impaired diabetic wound healing. The review also highlights the possible links between altered bone marrow myelopoiesis and increased Mo production as well as extrinsic and intrinsic factors that drive wound macrophage dysregulation leading to impaired wound healing in diabetes., (Copyright © 2020 Barman and Koh.)
- Published
- 2020
- Full Text
- View/download PDF
6. Heterogeneity of Mesenchymal Stromal Cells in Myelodysplastic Syndrome-with Multilineage Dysplasia (MDS-MLD).
- Author
-
Abbas S, Kumar S, Srivastava VM, Therese M M, Nair SC, Abraham A, Mathews V, George B, and Srivastava A
- Abstract
Bone marrow niche constituents have been implicated in the genesis of clonal hematopoietic dysfunction in myelodysplastic syndromes (MDS), though the exact role of stroma in the pathogenesis of MDS remains to be defined. We have evaluated the characteristics of mesenchymal stromal cells in a cohort of patients with MDS with multilineage dysplasia (MDS-MLD). MSCs were cultured from bone marrow aspirates of MDS-MLD patients and controls with healthy bone marrow. Phenotypic characterization, cell cycle, and apoptosis were analyzed by flow cytometry. Targeted gene expression analysis was done using a reverse-transcription polymerase chain reaction (Q-PCR). MSCs derived from MDS patients (MDS-MSCs) showed normal morphology, phenotype, karyotype and differentiation potential towards adipogenic and osteogenic lineages. However, these MDS-MSCs showed significantly altered cell cycle status and displayed a shift towards increased apoptosis compared to control MSCs (C-MSCs). The gene expression profile of niche responsive/regulatory cytokines showed a trend towards lower expression VEGF , SCF , and ANGPT with no changes in expression of CXCL12A and LIF compared to C-MSCs. The expression levels of Notch signaling components like Notch ligands ( JAGGED - 1 and DELTA - LIKE - 1 ), receptors ( NOTCH1 , NOTCH3 ) and downstream gene ( HES1 ) showed an aberrant expression pattern in MDS-MSCs compared to C-MSCs. Similarly, Q-PCR analysis of Wnt signaling inhibitory ligands ( DKK - 1 and DKK - 2 ) in MDS-MSCs showed a three-fold increase in mRNA expression of DKK1 and a two-fold increase in DKK2 compared to C-MSCs. These data suggested that MDS-MSCs have an altered proliferation characteristic as well as a dysregulated cytokine secretion and signaling profile. These changes could contribute to the pathogenesis of MDS., Competing Interests: Authors declares that they have no conflict of interest.All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.Informed consent was obtained from all individual participants included in the study.
- Published
- 2019
- Full Text
- View/download PDF
7. Superparamagnetic iron oxide nanoparticles for direct labeling of stem cells and in vivo MRI tracking.
- Author
-
Kim SJ, Lewis B, Steiner MS, Bissa UV, Dose C, and Frank JA
- Subjects
- Animals, Hematopoietic Stem Cells chemistry, Hematopoietic Stem Cells ultrastructure, Magnetic Resonance Imaging, Magnetite Nanoparticles chemistry, Mesenchymal Stem Cells chemistry, Mesenchymal Stem Cells ultrastructure, Mice, Microscopy, Electron, Transmission, Cell Proliferation, Cell Tracking, Ferric Compounds chemistry, Staining and Labeling
- Abstract
To develop effective stem cell therapies, it is important to track therapeutic cells non-invasively and monitor homing to areas of pathology. The purpose of this study was to design and evaluate the labeling efficiency of commercially available dextran-coated superparamagnetic iron oxide nanoparticles, FeraTrack Direct (FTD), in various stem and immune cells; assess the cytotoxicity and tolerability of the FTD in stem cells; and monitor stem cell homing using FTD-labeled bone-marrow-derived mesenchymal stromal cells (BMSCs) and neural stem cells (NSCs) in a tumor model by in vivo MRI. BMSCs, NSCs, hematopoietic stem cells (HSCs), T-lymphocytes, and monocytes were labeled effectively with FTD without the need for transfection agents, and Prussian blue (PB) staining and transmission electron microscopy (TEM) confirmed intracellular uptake of the agent. The viability, proliferation, and functionality of the labeled cells were minimally or not affected after labeling. When 10(6) FTD-labeled BMSCs or NSCs were injected into C6 glioma bearing nude mice, the cells homing to the tumors were detected as hypointense regions within the tumor using 3 T clinical MRI up to 10 days post injection. Histological analysis confirmed the homing of injected cells to the tumor by the presence of PB positive cells that are not macrophages. Labeling of stem cells or immune cells with FTD was non-toxic, and should facilitate the translation of this agent to clinical trials for evaluation of trafficking of cells by MRI., (Copyright © 2015 John Wiley & Sons, Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.