Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46.840
Filter
1.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727270

ABSTRACT

Self-renewal and differentiation are two characteristics of hematopoietic stem cells (HSCs). Under steady physiological conditions, most primitive HSCs remain quiescent in the bone marrow (BM). They respond to different stimuli to refresh the blood system. The transition from quiescence to activation is accompanied by major changes in metabolism, a fundamental cellular process in living organisms that produces or consumes energy. Cellular metabolism is now considered to be a key regulator of HSC maintenance. Interestingly, HSCs possess a distinct metabolic profile with a preference for glycolysis rather than oxidative phosphorylation (OXPHOS) for energy production. Byproducts from the cellular metabolism can also damage DNA. To counteract such insults, mammalian cells have evolved a complex and efficient DNA damage repair (DDR) system to eliminate various DNA lesions and guard genomic stability. Given the enormous regenerative potential coupled with the lifetime persistence of HSCs, tight control of HSC genome stability is essential. The intersection of DDR and the HSC metabolism has recently emerged as an area of intense research interest, unraveling the profound connections between genomic stability and cellular energetics. In this brief review, we delve into the interplay between DDR deficiency and the metabolic reprogramming of HSCs, shedding light on the dynamic relationship that governs the fate and functionality of these remarkable stem cells. Understanding the crosstalk between DDR and the cellular metabolism will open a new avenue of research designed to target these interacting pathways for improving HSC function and treating hematologic disorders.


Subject(s)
DNA Damage , DNA Repair , Hematopoietic Stem Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Animals , Genomic Instability , Energy Metabolism , Oxidative Phosphorylation
2.
Dev Cell ; 59(9): 1093-1095, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38714156

ABSTRACT

In this issue of Developmental Cell, Fowler et al. applied genetic lineage-tracing mouse models to support the notion that artery endothelial cells are the predominant source of hematopoietic stem cells. They leveraged this and developed a method capable of efficiently differentiating human pluripotent stem cells into HLF+HOXA+ hematopoietic progenitors.


Subject(s)
Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells , Pluripotent Stem Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Mice , Cell Lineage , Endothelial Cells/cytology , Endothelial Cells/metabolism
3.
Nat Commun ; 15(1): 3800, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714703

ABSTRACT

Clonal hematopoiesis (CH) is characterized by the acquisition of a somatic mutation in a hematopoietic stem cell that results in a clonal expansion. These driver mutations can be single nucleotide variants in cancer driver genes or larger structural rearrangements called mosaic chromosomal alterations (mCAs). The factors that influence the variations in mCA fitness and ultimately result in different clonal expansion rates are not well understood. We used the Passenger-Approximated Clonal Expansion Rate (PACER) method to estimate clonal expansion rate as PACER scores for 6,381 individuals in the NHLBI TOPMed cohort with gain, loss, and copy-neutral loss of heterozygosity mCAs. Our mCA fitness estimates, derived by aggregating per-individual PACER scores, were correlated (R2 = 0.49) with an alternative approach that estimated fitness of mCAs in the UK Biobank using population-level distributions of clonal fraction. Among individuals with JAK2 V617F clonal hematopoiesis of indeterminate potential or mCAs affecting the JAK2 gene on chromosome 9, PACER score was strongly correlated with erythrocyte count. In a cross-sectional analysis, genome-wide association study of estimates of mCA expansion rate identified a TCL1A locus variant associated with mCA clonal expansion rate, with suggestive variants in NRIP1 and TERT.


Subject(s)
Chromosome Aberrations , Clonal Hematopoiesis , Mosaicism , Humans , Clonal Hematopoiesis/genetics , Male , Female , Genome-Wide Association Study , Janus Kinase 2/genetics , Telomerase/genetics , Telomerase/metabolism , Loss of Heterozygosity , Cross-Sectional Studies , Mutation , Middle Aged , Hematopoietic Stem Cells/metabolism , Polymorphism, Single Nucleotide , Aged
4.
Gut Microbes ; 16(1): 2350784, 2024.
Article in English | MEDLINE | ID: mdl-38727219

ABSTRACT

The gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host's intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.


Subject(s)
Gastrointestinal Microbiome , Hematopoiesis , Hematopoietic Stem Cells , Hematopoiesis/physiology , Gastrointestinal Microbiome/physiology , Humans , Hematopoietic Stem Cells/microbiology , Animals , Signal Transduction , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Gastrointestinal Tract/microbiology , Bone Marrow/microbiology , Bone Marrow/physiology
5.
Hematology ; 29(1): 2347673, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38712914

ABSTRACT

The ability to perform hematopoietic cell transplant across major histocompatibility complex barriers can dramatically increase the availability of donors and allow more patients across the world to pursue curative transplant procedures for underlying hematologic disorders. Early attempts at haploidentical transplantation using broadly reactive T-cell depletion approaches were compromised by graft rejection, graft-versus-host disease and prolonged immune deficiency. The evolution of haploidentical transplantation focused on expanding transplanted hematopoietic progenitors as well as using less broadly reactive T-cell depletion. Significant outcome improvements were identified with technology advances allowing selective depletion of donor allospecific T cells, initially ex-vivo with evolution to its current in-vivo approach with the infusion of the highly immunosuppressive chemotherapy agent, cyclophosphamide after transplantation procedure. Current approaches are facile and portable, allowing expansion of allogeneic hematopoietic cell transplantation for patients across the world, including previously underserved populations.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation, Haploidentical , Humans , Hematopoietic Stem Cell Transplantation/methods , Transplantation, Haploidentical/methods , Graft vs Host Disease/prevention & control , Graft vs Host Disease/etiology , Hematopoietic Stem Cells/cytology
6.
Sci Adv ; 10(19): eadi6770, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38718114

ABSTRACT

Tracking stem cell fate transition is crucial for understanding their development and optimizing biomanufacturing. Destructive single-cell methods provide a pseudotemporal landscape of stem cell differentiation but cannot monitor stem cell fate in real time. We established a metabolic optical metric using label-free fluorescence lifetime imaging microscopy (FLIM), feature extraction and machine learning-assisted analysis, for real-time cell fate tracking. From a library of 205 metabolic optical biomarker (MOB) features, we identified 56 associated with hematopoietic stem cell (HSC) differentiation. These features collectively describe HSC fate transition and detect its bifurcate lineage choice. We further derived a MOB score measuring the "metabolic stemness" of single cells and distinguishing their division patterns. This score reveals a distinct role of asymmetric division in rescuing stem cells with compromised metabolic stemness and a unique mechanism of PI3K inhibition in promoting ex vivo HSC maintenance. MOB profiling is a powerful tool for tracking stem cell fate transition and improving their biomanufacturing from a single-cell perspective.


Subject(s)
Biomarkers , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells , Biomarkers/metabolism , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Cell Tracking/methods , Single-Cell Analysis/methods , Microscopy, Fluorescence/methods , Humans
7.
Blood ; 143(19): 1883-1884, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722657
8.
Cell Death Dis ; 15(5): 305, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693109

ABSTRACT

Zebrafish is widely adopted as a grafting model for studying human development and diseases. Current zebrafish xenotransplantations are performed using embryo recipients, as the adaptive immune system, responsible for host versus graft rejection, only reaches maturity at juvenile stage. However, transplanted primary human hematopoietic stem/progenitor cells (HSC) rapidly disappear even in zebrafish embryos, suggesting that another barrier to transplantation exists before the onset of adaptive immunity. Here, using a labelled macrophage zebrafish line, we demonstrated that engraftment of human HSC induces a massive recruitment of macrophages which rapidly phagocyte transplanted cells. Macrophages depletion, by chemical or pharmacological treatments, significantly improved the uptake and survival of transplanted cells, demonstrating the crucial implication of these innate immune cells for the successful engraftment of human cells in zebrafish. Beyond identifying the reasons for human hematopoietic cell engraftment failure, this work images the fate of human cells in real time over several days in macrophage-depleted zebrafish embryos.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Macrophages , Zebrafish , Zebrafish/embryology , Animals , Macrophages/metabolism , Humans , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cell Transplantation/methods , Embryo, Nonmammalian/metabolism , Transplantation, Heterologous , Phagocytosis
9.
J Clin Apher ; 39(3): e22114, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38708583

ABSTRACT

BACKGROUND: Stem cell apheresis in the context of autologous stem cell transplantation requires an accurate cluster of differentiantion 34 (CD34+) count determined by flow cytometry as the current gold standard. Since flow cytometry is a personnel and time-intensive diagnostic tool, automated stem cell enumeration may provide a promising alternative. Hence, this study aimed to compare automated hematopoietic progenitor enumeration carried out on a Sysmex XN-20 module compared with conventional flow cytometric measurements. METHODS: One hundred forty-three blood samples from 41 patients were included in this study. Correlation between the two methods was calculated over all samples, depending on leukocyte count and diagnosis. RESULTS: Overall, we found a high degree of correlation (r = 0.884). Furthermore, correlation was not impaired by elevated leukocyte counts (>10 000/µL, r = 0.860 vs <10 000/µL, r = 0.849; >20 000/µL, r = 0.843 vs <20 000/µL, r = 0.875). However, correlation was significantly impaired in patients with multiple myeloma (multiple myeloma r = 0.840 vs nonmyeloma r = 0.934). SUMMARY: Stem cell measurement carried out on the Sysmex XN-20 module provides a significant correlation with flow cytometry and might be implemented in clinical practice. In clinical decision-making, there was discrepancy of under 15% of cases. In multiple myeloma patients, XN-20 should be used with caution.


Subject(s)
Antigens, CD34 , Flow Cytometry , Hematopoietic Stem Cells , Humans , Flow Cytometry/methods , Antigens, CD34/analysis , Antigens, CD34/blood , Hematopoietic Stem Cells/cytology , Multiple Myeloma/blood , Multiple Myeloma/diagnosis , Male , Female , Blood Cell Count/methods , Blood Cell Count/instrumentation , Middle Aged , Leukocyte Count , Adult
10.
Sci Immunol ; 9(95): eadq0013, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701191

ABSTRACT

Antibody-based therapy depletes myeloid-biased hematopoietic stem cells (my-HSCs) to rejuvenate the immune system and improve immune responses in aged mice.


Subject(s)
Aging , Hematopoietic Stem Cells , Animals , Aging/immunology , Hematopoietic Stem Cells/immunology , Mice , Humans , Hematopoietic Stem Cell Transplantation
11.
Stem Cell Res Ther ; 15(1): 133, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704588

ABSTRACT

BACKGROUND: Human hematopoietic organoids have a wide application value for modeling human bone marrow diseases, such as acute hematopoietic radiation injury. However, the manufacturing of human hematopoietic organoids is an unaddressed challenge because of the complexity of hematopoietic tissues. METHODS: To manufacture hematopoietic organoids, we obtained CD34+ hematopoietic stem and progenitor cells (HSPCs) from human embryonic stem cells (hESCs) using stepwise induction and immunomagnetic bead-sorting. We then mixed these CD34+ HSPCs with niche-related cells in Gelatin-methacryloyl (GelMA) to form a three-dimensional (3D) hematopoietic organoid. Additionally, we investigated the effects of radiation damage and response to granulocyte colony-stimulating factor (G-CSF) in hematopoietic organoids. RESULTS: The GelMA hydrogel maintained the undifferentiated state of hESCs-derived HSPCs by reducing intracellular reactive oxygen species (ROS) levels. The established hematopoietic organoids in GelMA with niche-related cells were composed of HSPCs and multilineage blood cells and demonstrated the adherence of hematopoietic cells to niche cells. Notably, these hematopoietic organoids exhibited radiation-induced hematopoietic cell injury effect, including increased intracellular ROS levels, γ-H2AX positive cell percentages, and hematopoietic cell apoptosis percentages. Moreover, G-CSF supplementation in the culture medium significantly improved the survival of HSPCs and enhanced myeloid cell regeneration in these hematopoietic organoids after radiation. CONCLUSIONS: These findings substantiate the successful manufacture of a preliminary 3D hematopoietic organoid from hESCs-derived HSPCs, which was utilized for modeling hematopoietic radiation injury and assessing the radiation-mitigating effects of G-CSF in vitro. Our study provides opportunities to further aid in the standard and scalable production of hematopoietic organoids for disease modeling and drug testing.


Subject(s)
Granulocyte Colony-Stimulating Factor , Hematopoietic Stem Cells , Organoids , Humans , Organoids/metabolism , Organoids/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/cytology , Granulocyte Colony-Stimulating Factor/pharmacology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Regeneration/drug effects , Cell Differentiation/drug effects , Antigens, CD34/metabolism
12.
Sci Transl Med ; 16(745): eadi8214, 2024 May.
Article in English | MEDLINE | ID: mdl-38691622

ABSTRACT

Mucopolysaccharidosis type I Hurler (MPSIH) is characterized by severe and progressive skeletal dysplasia that is not fully addressed by allogeneic hematopoietic stem cell transplantation (HSCT). Autologous hematopoietic stem progenitor cell-gene therapy (HSPC-GT) provides superior metabolic correction in patients with MPSIH compared with HSCT; however, its ability to affect skeletal manifestations is unknown. Eight patients with MPSIH (mean age at treatment: 1.9 years) received lentiviral-based HSPC-GT in a phase 1/2 clinical trial (NCT03488394). Clinical (growth, measures of kyphosis and genu velgum), functional (motor function, joint range of motion), and radiological [acetabular index (AI), migration percentage (MP) in hip x-rays and MRIs and spine MRI score] parameters of skeletal dysplasia were evaluated at baseline and multiple time points up to 4 years after treatment. Specific skeletal measures were retrospectively compared with an external cohort of HSCT-treated patients. At a median follow-up of 3.78 years after HSPC-GT, all patients treated with HSPC-GT exhibited longitudinal growth within WHO reference ranges and a median height gain greater than that observed in patients treated with HSCT after 3-year follow-up. Patients receiving HSPC-GT experienced complete and earlier normalization of joint mobility compared with patients treated with HSCT. Mean AI and MP showed progressive decreases after HSPC-GT, suggesting a reduction in acetabular dysplasia. Typical spine alterations measured through a spine MRI score stabilized after HSPC-GT. Clinical, functional, and radiological measures suggested an early beneficial effect of HSPC-GT on MPSIH-typical skeletal features. Longer follow-up is needed to draw definitive conclusions on HSPC-GT's impact on MPSIH skeletal dysplasia.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Mucopolysaccharidosis I , Humans , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis I/genetics , Male , Female , Child, Preschool , Infant , Treatment Outcome , Hematopoietic Stem Cells/metabolism , Child , Bone and Bones/pathology , Magnetic Resonance Imaging
13.
Exp Clin Transplant ; 22(3): 229-238, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38695592

ABSTRACT

OBJECTIVES: The eradication of leukemia cells while sparing hematopoietic stem cells in the graft before autologous hematopoietic stem cell transplant is critical to prevention of leukemia relapse. Proliferating cells have been shown to be more prone to apoptosis than differentiated cells in response to ultraviolet radiation; however, whether leukemia cells are more sensitive to ultraviolet LED radiation than hematopoietic stem cells remains unclear. MATERIALS AND METHODS: We compared the in vitro responses between murine leukemia L1210 cells and murine hematopoietic stem cells to 280-nm ultraviolet LED radiation. We also investigated the effects of ultraviolet LED radiation on the tumorigenic and metastatic capacity of L1210 cells and hematopoietic stem cell hematopoiesis in a mouse model of hematopoietic stem cell transplant. RESULTS: L1210 cells were more sensitive to ultraviolet LED radiation than hematopoietic stem cells in vitro, as evidenced by significantly reduced colony formation rates and cell proliferation rates, along with remarkably increased apoptosis rates in L1210 cells. Compared with corresponding unirradiated cells, ultraviolet LED-irradiated L1210 cells failed to generate palpable tumors in mice, whereas ultraviolet LED-irradiated bone marrow cells restored hematopoiesis in vivo. Furthermore, transplant with an irradiated mixture of L1210 cells and bone marrow cells showed later onset of leukemia, milder leukemic infiltration, and prolonged survival in mice, compared with unirradiated cell transplant. CONCLUSIONS: Our results suggest that ultraviolet LED radiation can suppress the proliferative and tumorigenic abilities of leukemia cells without reducing the hematopoietic reconstitution capacity of hematopoietic stem cells, serving as a promising approach to kill leukemia cells in autograft before autologous hematopoietic stem cell transplant.


Subject(s)
Apoptosis , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/radiation effects , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/metabolism , Apoptosis/radiation effects , Hematopoiesis/radiation effects , Cell Proliferation/radiation effects , Cell Line, Tumor , Ultraviolet Rays/adverse effects , Mice , Mice, Inbred C57BL , Time Factors , Ultraviolet Therapy
15.
Blood ; 143(16): 1554-1556, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38635250
16.
J Vis Exp ; (205)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38558008

ABSTRACT

Induced pluripotent stem cell (iPSC)-based models are excellent platforms to understand blood development, and iPSC-derived blood cells have translational utility as clinical testing reagents and transfusable cell therapeutics. The advent and expansion of multiomics analysis, including but not limited to single nucleus RNA sequencing (snRNAseq) and Assay for Transposase-Accessible Chromatin sequencing (snATACseq), offers the potential to revolutionize our understanding of cell development. This includes developmental biology using in vitro hematopoietic models. However, it can be technically challenging to isolate intact nuclei from cultured or primary cells. Different cell types often require tailored nuclear preparations depending on cellular rigidity and content. These technical difficulties can limit data quality and act as a barrier to investigators interested in pursuing multiomics studies. Specimen cryopreservation is often necessary due to limitations with cell collection and/or processing, and frozen samples can present additional technical challenges for intact nuclear isolation. In this manuscript, we provide a detailed method to isolate high-quality nuclei from iPSC-derived cells at different stages of in vitro hematopoietic development for use in single-nucleus multiomics workflows. We have focused the method development on the isolation of nuclei from iPSC-derived adherent stromal/endothelial cells and non-adherent hematopoietic progenitor cells, as these represent very different cell types with regard to structural and cellular identity. The described troubleshooting steps limited nuclear clumping and debris, allowing the recovery of nuclei in sufficient quantity and quality for downstream analyses. Similar methods may be adapted to isolate nuclei from other cryopreserved cell types.


Subject(s)
Cell Nucleus , Endothelial Cells , Cell Nucleus/metabolism , Cryopreservation/methods , Hematopoietic Stem Cells , Blood Cells
17.
Transfusion ; 64(5): 866-870, 2024 May.
Article in English | MEDLINE | ID: mdl-38606842

ABSTRACT

BACKGROUND: Standard flow cytometry protocols for CD34+ cell enumeration designed for fresh samples are not appropriate for cryopreserved products. Special protocols have been developed to remove the cryoprotectant by quickly washing a freshly thawed sample. Exposing cells to a large volume of hypotonic solution and subsequent washing process was hypothesized to cause lab-induced cell death. Moreover, standard gating strategies must be altered to avoid reporting falsely high viabilities. STUDY DESIGN AND METHODS: We developed a novel method whereby thawed samples were diluted step-wise to 1:2 by 3 additions of 1/3 sample volume using 1% Human Albumin in Dextran 40 (10% Low Molecular Weight Dextran in 0.9% NaCl) separated by 5 min between each addition. An additional 1:10 dilution was required to obtain a desired cell concentration for flow cytometry testing resulting in a 1:20 dilution. RESULTS: Twenty samples were tested simultaneously in a method comparison; the new method demonstrated significant increases in mean cell viabilities for white blood cells, hematopoietic progenitor cells, and T cells as well as reduced standard deviations for each parameter. DISCUSSION: Slow, step-wise dilutions of freshly thawed samples of cryopreserved apheresis products to 1:20 yielded higher and more precise viability measurements compared to quickly washing samples to remove DMSO.


Subject(s)
Blood Component Removal , Cell Survival , Cryopreservation , Flow Cytometry , Humans , Cryopreservation/methods , Flow Cytometry/methods , Blood Component Removal/methods , Hematopoietic Stem Cells/cytology , Blood Preservation/methods , Cryoprotective Agents/pharmacology , Antigens, CD34/analysis
18.
Aging (Albany NY) ; 16(7): 5796-5810, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38604248

ABSTRACT

Despite their biological importance, the role of stem cells in human aging remains to be elucidated. In this work, we applied a machine learning methodology to GTEx transcriptome data and assigned stemness scores to 17,382 healthy samples from 30 human tissues aged between 20 and 79 years. We found that ~60% of the studied tissues exhibit a significant negative correlation between the subject's age and stemness score. The only significant exception was the uterus, where we observed an increased stemness with age. Moreover, we observed that stemness is positively correlated with cell proliferation and negatively correlated with cellular senescence. Finally, we also observed a trend that hematopoietic stem cells derived from older individuals might have higher stemness scores. In conclusion, we assigned stemness scores to human samples and show evidence of a pan-tissue loss of stemness during human aging, which adds weight to the idea that stem cell deterioration may contribute to human aging.


Subject(s)
Aging , Cellular Senescence , Humans , Aging/physiology , Aged , Middle Aged , Adult , Female , Cellular Senescence/physiology , Stem Cells/metabolism , Male , Cell Proliferation , Young Adult , Transcriptome , Machine Learning , Hematopoietic Stem Cells/metabolism
19.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667319

ABSTRACT

Platelets are the terminal progeny of megakaryocytes, primarily produced in the bone marrow, and play critical roles in blood homeostasis, clotting, and wound healing. Traditionally, megakaryocytes and platelets are thought to arise from multipotent hematopoietic stem cells (HSCs) via multiple discrete progenitor populations with successive, lineage-restricting differentiation steps. However, this view has recently been challenged by studies suggesting that (1) some HSC clones are biased and/or restricted to the platelet lineage, (2) not all platelet generation follows the "canonical" megakaryocytic differentiation path of hematopoiesis, and (3) platelet output is the default program of steady-state hematopoiesis. Here, we specifically investigate the evidence that in vivo lineage tracing studies provide for the route(s) of platelet generation and investigate the involvement of various intermediate progenitor cell populations. We further identify the challenges that need to be overcome that are required to determine the presence, role, and kinetics of these possible alternate pathways.


Subject(s)
Blood Platelets , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Blood Platelets/cytology , Blood Platelets/metabolism , Mice , Megakaryocytes/cytology , Megakaryocytes/metabolism , Hematopoiesis
20.
Blood Cancer Discov ; 5(3): 139-141, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38651690

ABSTRACT

SUMMARY: The spatial distribution of cells carrying clonal hematopoiesis mutations in the bone marrow and the potential role of interactions with the microenvironment are largely unknown. This study takes clonal evolution to the spatial level by describing a novel technique examining the spatial location of mutated clones in the bone marrow and the first evidence that mutated hematopoietic clones are spatially constrained and have heterogenous locations within millimeters of distance. See related article by Young et al., p. 153 (10).


Subject(s)
Clonal Evolution , Clonal Hematopoiesis , Mutation , Clonal Evolution/genetics , Humans , Clonal Hematopoiesis/genetics , Bone Marrow , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...