Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47.058
Filter
1.
Stem Cell Res Ther ; 15(1): 248, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113086

ABSTRACT

BACKGROUND: The function of hematopoietic stem cells (HSC) is regulated by HSC internal signaling pathways and their microenvironment. Chemokines and chemokine ligands play important roles in the regulation of HSC function. Yet, their functions in HSC are not fully understood. METHODS: We established Cxcr3 and Cxcl10 knockout mouse models (Cxcr3-/- and Cxcl10-/-) to analyze the roles of Cxcr3 or Cxcl10 in regulating HSC function. The cell cycle distribution of LT-HSC was assessed via flow cytometry. Cxcr3-/- and Cxcl10-/- stem/progenitor cells showed reduced self-renewal capacity as measured in serial transplantation assays. To study the effects of Cxcr3 or Cxcl10 deficient bone marrow microenvironment, we transplanted CD45.1 donor cells into Cxcr3-/-or Cxcl10-/- recipient mice (CD45.2) and examined donor-contributed hematopoiesis. RESULTS: Deficiency of Cxcl10 and its receptor Cxcr3 led to decreased BM cellularity in mice, with a significantly increased proportion of LT-HSC. Cxcl10-/- stem/progenitor cells showed reduced self-renewal capacity in the secondary transplantation assay. Notably, Cxcl10-/- donor-derived cells preferentially differentiated into B lymphocytes, with skewed myeloid differentiation ability. Meanwhile, Cxcr3-deficient HSCs demonstrated a reconstitution disadvantage in secondary transplantation, but the lineage bias was not significant. Interestingly, the absence of Cxcl10 or Cxcr3 in bone marrow microenvironment did not affect HSC function. CONCLUSIONS: The Cxcl10 and Cxcr3 regulate the function of HSC, including self-renewal and differentiation, adding to the understanding of the roles of chemokines in the regulation of HSC function.


Subject(s)
Cell Differentiation , Chemokine CXCL10 , Hematopoietic Stem Cells , Receptors, CXCR3 , Animals , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Mice, Inbred C57BL , Cell Self Renewal , Hematopoiesis , Hematopoietic Stem Cell Transplantation
2.
Clin Adv Hematol Oncol ; 22(7): 320-327, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39110659

ABSTRACT

As individuals age, their hematopoietic stem cells can sporadically acquire genetic mutations, known as clonal hematopoiesis. Although most of these genomic aberrations are of little consequence, particular changes in certain contexts can lead to the development of hematologic malignancies, such as myelodysplastic syndromes and acute myeloid leukemia. Owing to its pervasive extrahematologic interactions, clonal hematopoiesis is a recognized risk factor for and is causally implicated in the development of several chronic diseases of aging and/or inflammation, such as atherosclerotic cardiovascular disease. Here, we provide a review of the diagnosis and clinical implications of clonal hematopoiesis, as well as evolving management strategies in the absence of formal consensus guidelines.


Subject(s)
Clonal Hematopoiesis , Humans , Myelodysplastic Syndromes/therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/etiology , Mutation , Disease Management , Hematologic Neoplasms/therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/diagnosis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology
3.
Handb Clin Neurol ; 202: 105-115, 2024.
Article in English | MEDLINE | ID: mdl-39111903

ABSTRACT

Most hematopoietic stem cell transplants performed for an autoimmune disease of the nervous system, use the patient's hematopoietic stem cells (HSCs). Obtaining an HSC graft is the first step of the process. This typically involves mobilization of bone marrow HSCs into the circulation using high-dose cyclophosphamide followed by filgrastim, a drug based on granulocyte colony-stimulating factor. Toxicity from these agents is usually manageable and adverse events are less severe and less frequent than those experienced during the hematopoietic stem cell transplant. Following mobilization, HSCs are collected from the circulation by leukapheresis. Some centers deplete the graft of lymphocytes using an ex vivo immunomagnetic selection procedure. HSC grafts are cryopreserved until required for the stem cell transplant. Quality testing of the graft ensures sterility and it contains sufficient HSCs and hematopoietic progenitors. The clinical and laboratory aspects of HSC graft mobilization, collection, and storage must meet standards set by national regulatory bodies and accredited by international professional standards organizations. Experienced stem cell transplant teams are important for minimizing procedural toxicity and enhancing successful collection.


Subject(s)
Cryopreservation , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Humans , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Cryopreservation/methods , Hematopoietic Stem Cells
4.
Handb Clin Neurol ; 202: 23-39, 2024.
Article in English | MEDLINE | ID: mdl-39111909

ABSTRACT

Hematopoiesis is a complex process that takes place inside the bone marrow, where a specialized structure, the bone marrow niche, participates in the maintenance of hematopoietic stem cell functionality. Inflammatory conditions, such as autoimmune diseases, could alter this equilibrium leading to pathologic consequences. Immune cells, which also reside in the bone marrow, directly participate in sustaining the inflammatory state in autoimmune diseases. In particular, memory lymphocytes are key players in the long-term maintenance of the immune response against self-antigens, causing tissue damage and bone marrow alterations.


Subject(s)
Autoimmune Diseases , Humans , Autoimmune Diseases/immunology , Animals , Immunologic Memory/immunology , Hematopoiesis/physiology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology
5.
Nat Commun ; 15(1): 6787, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117713

ABSTRACT

The maintenance of hematopoietic stem cell (HSC) functional integrity is essential for effective hematopoietic regeneration when suffering from injuries. Studies have shown that the innate immune pathways play crucial roles in the stress response of HSCs, whereas how to precisely modulate these pathways is not well characterized. Here, we identify the E3 ubiquitin ligase tripartite motif-containing 47 (Trim47) as a negative regulator of the mitochondrial antiviral-signaling protein (MAVS)-mediated innate immune pathway in HSCs. We find that Trim47 is predominantly enriched in HSCs, and its deficiency impairs the function and survival of HSCs after exposure to 5-flurouracil (5-FU) and irradiation (IR). Mechanistically, Trim47 impedes the excessive activation of the innate immune signaling and inflammatory response via K48-linked ubiquitination and degradation of MAVS. Collectively, our findings demonstrate a role of Trim47 in preventing stress-induced hematopoietic failure and thus provide a promising avenue for treatment of related diseases in the clinic.


Subject(s)
Adaptor Proteins, Signal Transducing , Hematopoietic Stem Cells , Immunity, Innate , Mice, Inbred C57BL , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , Animals , Hematopoietic Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mice, Knockout , Humans , Fluorouracil/pharmacology , Stress, Physiological , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , HEK293 Cells
6.
Commun Biol ; 7(1): 967, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122870

ABSTRACT

The mitochondrial permeability transition pore (mPTP) is a supramolecular channel that regulates exchange of solutes across cristae membranes, with executive roles in mitochondrial function and cell death. The contribution of the mPTP to normal physiology remains debated, although evidence implicates the mPTP in mitochondrial inner membrane remodeling in differentiating progenitor cells. Here, we demonstrate that strict control over mPTP conductance shapes metabolic machinery as cells transit toward hematopoietic identity. Cells undergoing the endothelial-to-hematopoietic transition (EHT) tightly control chief regulatory elements of the mPTP. During EHT, maturing arterial endothelium restricts mPTP activity just prior to hematopoietic commitment. After transition in cellular identity, mPTP conductance is restored. In utero treatment with NIM811, a molecule that blocks sensitization of the mPTP to opening by Cyclophilin D (CypD), amplifies oxidative phosphorylation (OXPHOS) in hematopoietic precursors and increases hematopoiesis in the embryo. Additionally, differentiating pluripotent stem cells (PSCs) acquire greater organization of mitochondrial cristae and hematopoietic activity following knockdown of the CypD gene, Ppif. Conversely, knockdown of Opa1, a GTPase critical for proper cristae architecture, induces cristae irregularity and impairs hematopoiesis. These data elucidate a mechanism that regulates mitochondrial maturation in hematopoietic precursors and underscore a role for the mPTP in the acquisition of hematopoietic fate.


Subject(s)
Hematopoietic Stem Cells , Mitochondria , Mitochondrial Permeability Transition Pore , Animals , Mitochondria/metabolism , Hematopoietic Stem Cells/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Hematopoiesis , Peptidyl-Prolyl Isomerase F/metabolism , Peptidyl-Prolyl Isomerase F/genetics , Cell Differentiation , Oxidative Phosphorylation , Female , Mice, Inbred C57BL
7.
BMC Genomics ; 25(1): 782, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134978

ABSTRACT

BACKGROUND: The Lin-Sca1+c-Kit+ (LSK) fraction of the bone marrow (BM) comprises multipotent hematopoietic stem cells (HSCs), which are vital to tissue homeostasis and vascular repair. While diabetes affects HSC homeostasis overall, the molecular signature of mRNA and miRNA transcriptomic under the conditions of long-standing type 2 diabetes (T2D;>6 months) remains unexplored. METHODS: In this study, we assessed the transcriptomic signature of HSCs in db/db mice, a well-known and widely used model for T2D. LSK cells of db/db mice enriched using a cell sorter were subjected to paired-end mRNA and single-end miRNA seq library and sequenced on Illumina NovaSeq 6000. The mRNA sequence reads were mapped using STAR (Spliced Transcripts Alignment to a Reference), and the miRNA sequence reads were mapped to the designated reference genome using the Qiagen GeneGlobe RNA-seq Analysis Portal with default parameters for miRNA. RESULTS: We uncovered 2076 out of 13,708 mRNAs and 35 out of 191 miRNAs that were expressed significantly in db/db animals; strikingly, previously unreported miRNAs (miR-3968 and miR-1971) were found to be downregulated in db/db mice. Furthermore, we observed a molecular shift in the transcriptome of HSCs of diabetes with an increase in pro-inflammatory cytokines (Il4, Tlr4, and Tnf11α) and a decrease in anti-inflammatory cytokine IL10. Pathway mapping demonstrated inflammation mediated by chemokine, cytokine, and angiogenesis as one of the top pathways with a significantly higher number of transcripts in db/db mice. These molecular changes were reflected in an overt defect in LSK mobility in the bone marrow. miRNA downstream target analysis unveils several mRNAs targeting leukocyte migration, microglia activation, phagosome formation, and macrophage activation signaling as their primary pathways, suggesting a shift to an inflammatory phenotype. CONCLUSION: Our findings highlight that chronic diabetes adversely alters HSCs' homeostasis at the transcriptional level, thus potentially contributing to the inflammatory phenotype of HSCs under long-term diabetes. We also believe that identifying HSCs-based biomarkers in miRNAs or mRNAs could serve as diagnostic markers and potential therapeutic targets for diabetes and associated vascular complications.


Subject(s)
Diabetes Mellitus, Type 2 , Hematopoietic Stem Cells , MicroRNAs , Transcriptome , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Hematopoietic Stem Cells/metabolism , Gene Expression Profiling , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Male , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism
10.
Adv Gerontol ; 37(3): 266-275, 2024.
Article in Russian | MEDLINE | ID: mdl-39139119

ABSTRACT

The number of somatic mutations among all tissues increases along with age. This process was well-studied in hematopoietic stem cells (HSCs). Some mutations lead to a proliferative advantage and expansion of HSCs to form a dominant clone. Clonal hematopoiesis is general in the elderly population. Clonal hematopoiesis of indeterminate potential (CHIP) is a more common phenomenon in the elderly and is defined as somatic mutations in clonal blood cells without any other hematological malignancies. The development of CHIP is an independent risk factor for hematological malignancies, cardiovascular diseases, and reduced overall survival. CHIP is frequently associated with mutations in DNMT3A and TET2 genes involved in DNA methylation. The epigenetic human body clocks have been developed based on the age-related changes in methylation, making it possible to detect epigenetic aging. The combination of epigenetic aging and CHUP is associated with adverse health outcomes. Further research will reveal the significance of clonal hematopoiesis and CHIP in aging, acquiring various diseases, and determining the feasibility of influencing the mutagenic potential of clones.


Subject(s)
Aging , Clonal Hematopoiesis , Epigenesis, Genetic , Humans , Aging/physiology , Aging/genetics , Clonal Hematopoiesis/genetics , Mutation , DNA Methylation , Hematopoietic Stem Cells/metabolism , DNA Methyltransferase 3A , Hematologic Neoplasms/genetics , Hematologic Neoplasms/diagnosis , Dioxygenases , Hematopoiesis/genetics , Hematopoiesis/physiology , DNA (Cytosine-5-)-Methyltransferases/genetics
11.
Front Immunol ; 15: 1365946, 2024.
Article in English | MEDLINE | ID: mdl-39131155

ABSTRACT

Introduction: Humanized mouse models to recapitulate human biological systems still have limitations, such as the onset of lethal graft-versus-host disease (GvHD), a variable success rate, and the low accessibility of total body irradiation (TBI). Recently, mice modified with the CD47-SIRPA axis have been studied to improve humanized mouse models. However, such trials have been rarely applied in NOD mice. In this study, we created a novel mouse strain, NOD-CD47nullRag2nullIL-2rγnull (RTKO) mice, and applied it to generate humanized mice. Methods: Four-week-old female NOD-Rag2nullIL-2rγnull (RID) and RTKO mice pre-conditioned with TBI or busulfan (BSF) injection were used for generating human CD34+ hematopoietic stem cell (HSC) engrafted humanized mice. Clinical signs were observed twice a week, and body weight was measured once a week. Flow cytometry for human leukocyte antigens was performed at intervals of four weeks or two weeks, and mice were sacrificed at 48 weeks after HSC injection. Results: For a long period from 16 to 40 weeks post transplantation, the percentage of hCD45 was mostly maintained above 25% in all groups, and it was sustained the longest and highest in the RTKO BSF group. Reconstruction of human leukocytes, including hCD3, was also most prominent in the RTKO BSF group. Only two mice died before 40 weeks post transplantation in all groups, and there were no life-threatening GvHD lesions except in the dead mice. The occurrence of GvHD has been identified as mainly due to human T cells infiltrating tissues and their related cytokines. Discussion: Humanized mouse models under all conditions applied in this study are considered suitable models for long-term experiments based on the improvement of human leukocytes reconstruction and the stable animal health. Especially, RTKO mice pretreated with BSF are expected to be a valuable platform not only for generating humanized mice but also for various immune research fields.


Subject(s)
Busulfan , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Mice, Inbred NOD , Mice, Knockout , Transplantation Conditioning , Animals , Busulfan/pharmacology , Humans , Mice , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Hematopoietic Stem Cells/metabolism , Female , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/deficiency , Graft vs Host Disease/prevention & control , Graft vs Host Disease/immunology , Disease Models, Animal , Whole-Body Irradiation
12.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087468

ABSTRACT

Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by the selective expansion of hematopoietic stem and progenitor cells (HSPCs) carrying somatic mutations. While CHIP is typically asymptomatic, it has garnered substantial attention due to its association with the pathogenesis of multiple disease conditions, including cardiovascular disease (CVD) and hematological malignancies. In this Review, we will discuss seminal and recent studies that have advanced our understanding of mechanisms that drive selection for mutant HSPCs in the BM niche. Next, we will address recent studies evaluating potential relationships between the clonal dynamics of CHIP and hematopoietic development across the lifespan. Next, we will examine the roles of systemic factors that can influence hematopoietic stem cell (HSC) fitness, including inflammation, and exposures to cytotoxic agents in driving selection for CHIP clones. Furthermore, we will consider how - through their impact on the BM niche - lifestyle factors, including diet, exercise, and psychosocial stressors, might contribute to the process of somatic evolution in the BM that culminates in CHIP. Finally, we will review the role of old age as a major driver of selection in CHIP.


Subject(s)
Clonal Hematopoiesis , Hematopoietic Stem Cells , Stem Cell Niche , Humans , Hematopoietic Stem Cells/metabolism , Clonal Hematopoiesis/genetics , Animals , Bone Marrow/metabolism , Mutation
13.
Science ; 385(6709): eadp2065, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39116219

ABSTRACT

Hematopoietic stem cells (HSCs) are routinely mobilized from the bone marrow (BM) to the blood circulation for clinical transplantation. However, the precise mechanisms by which individual stem cells exit the marrow are not understood. This study identified cell-extrinsic and molecular determinants of a mobilizable pool of blood-forming stem cells. We found that a subset of HSCs displays macrophage-associated markers on their cell surface. Although fully functional, these HSCs are selectively niche-retained as opposed to stem cells lacking macrophage markers, which exit the BM upon forced mobilization. Macrophage markers on HSCs could be acquired through direct transfer by trogocytosis, regulated by receptor tyrosine-protein kinase C-Kit (CD117), from BM-resident macrophages in mouse and human settings. Our study provides proof of concept that adult stem cells utilize trogocytosis to rapidly establish and activate function-modulating molecular mechanisms.


Subject(s)
Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells , Proto-Oncogene Proteins c-kit , Trogocytosis , Animals , Humans , Mice , Adult Stem Cells/physiology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Macrophages/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-kit/genetics , Stem Cell Niche , Sialic Acid Binding Ig-like Lectin 1/metabolism , Antigens, Differentiation
14.
Stem Cell Res Ther ; 15(1): 251, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135194

ABSTRACT

Collection of hematopoietic progenitor cell products [HPC(A)] is deferred if the donor is symptomatic and tests positive for Covid-19. However, donor questionnaires are subjective and may miss minimally symptomatic donors. Alternatively, myalgia associated with Covid-19 infection can be falsely dismissed as an adverse effect of granulocyte stimulating factor (Filgrastim) administered prior to product collection. The likelihood of donors with an underlying acute but minimally symptomatic infection undergoing successful product collection is significant. In these circumstances, it is less known whether Covid-19 infection results in product viremia or alters the clinical outcome of transplant. We aimed to evaluate the above question by studying a donor whose product was collected during acute Covid-19 infection. Aliquots of the product tested negative for SARS-CoV-2 RNA by reverse-transcriptase polymerase chain reaction assay (RT-PCR). Importantly, the donor received an autologous stem cell transplant using the product collected at the time of infection, and their case will be described in this report. We describe one of the very few reports of successful transplant of HPC(A) product collected during acute Covid-19 infection.


Subject(s)
COVID-19 , Hematopoietic Stem Cell Transplantation , SARS-CoV-2 , Transplantation, Autologous , Humans , COVID-19/virology , COVID-19/therapy , SARS-CoV-2/genetics , Male , Middle Aged , Tissue Donors , Female , Adult , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/virology
15.
Stem Cell Reports ; 19(8): 1189-1204, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094562

ABSTRACT

It has been proposed that adult hematopoiesis is sustained by multipotent progenitors (MPPs) specified during embryogenesis. Adult-like hematopoietic stem cell (HSC) and MPP immunophenotypes are present in the fetus, but knowledge of their functional capacity is incomplete. We found that fetal MPP populations were functionally similar to adult cells, albeit with some differences in lymphoid output. Clonal assessment revealed that lineage biases arose from differences in patterns of single-/bi-lineage differentiation. Long-term (LT)- and short-term (ST)-HSC populations were distinguished from MPPs according to capacity for clonal multilineage differentiation. We discovered that a large cohort of long-term repopulating units (LT-RUs) resides within the ST-HSC population; a significant portion of these were labeled using Flt3-cre. This finding has two implications: (1) use of the CD150+ LT-HSC immunophenotype alone will significantly underestimate the size and diversity of the LT-RU pool and (2) LT-RUs in the ST-HSC population have the attributes required to persist into adulthood.


Subject(s)
Cell Lineage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Cell Differentiation , Fetus/cytology , Immunophenotyping , Hematopoiesis , Clone Cells/cytology
16.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39171985

ABSTRACT

The tendency for cell fate to be robust to most perturbations, yet sensitive to certain perturbations raises intriguing questions about the existence of a key path within the underlying molecular network that critically determines distinct cell fates. Reprogramming and trans-differentiation clearly show examples of cell fate change by regulating only a few or even a single molecular switch. However, it is still unknown how to identify such a switch, called a master regulator, and how cell fate is determined by its regulation. Here, we present CAESAR, a computational framework that can systematically identify master regulators and unravel the resulting canalizing kernel, a key substructure of interconnected feedbacks that is critical for cell fate determination. We demonstrate that CAESAR can successfully predict reprogramming factors for de-differentiation into mouse embryonic stem cells and trans-differentiation of hematopoietic stem cells, while unveiling the underlying essential mechanism through the canalizing kernel. CAESAR provides a system-level understanding of how complex molecular networks determine cell fates.


Subject(s)
Cell Differentiation , Animals , Mice , Cellular Reprogramming , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Computational Biology/methods , Gene Regulatory Networks , Cell Lineage , Cell Transdifferentiation
17.
Sci Immunol ; 9(98): eadk3469, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178276

ABSTRACT

Hematopoietic stem cells (HSCs) reconstitute multilineage human hematopoiesis after clinical bone marrow (BM) transplantation and are the cells of origin of some hematological malignancies. Although HSCs provide multilineage engraftment, individual murine HSCs are lineage biased and contribute unequally to blood cell lineages. Here, we performed high-throughput single-cell RNA sequencing in mice after xenograft with molecularly barcoded adult human BM HSCs. We demonstrated that human individual BM HSCs are also functionally and transcriptionally lineage biased. Specifically, we identified platelet-biased and multilineage human HSCs. Quantitative comparison of transcriptomes from single HSCs from young and aged BM showed that both the proportion of platelet-biased HSCs and their level of transcriptional platelet priming increase with age. Therefore, platelet-biased HSCs and their increased prevalence and transcriptional platelet priming during aging are conserved features of mammalian evolution.


Subject(s)
Blood Platelets , Hematopoietic Stem Cells , Animals , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/cytology , Humans , Mice , Blood Platelets/immunology , Aging/immunology , Cell Lineage/immunology , Biological Evolution , Single-Cell Analysis
18.
FASEB J ; 38(15): e23870, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39120151

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) are successfully employed for hematological transplantations, and impaired HSPC function causes hematological diseases and aging. HSPCs maintain the lifelong homeostasis of blood and immune cells through continuous self-renewal and maintenance of the multilineage differentiation potential. TMEM106B is a transmembrane protein localized on lysosomal membranes and associated with neurodegenerative and cardiovascular diseases; however, its roles in HSPCs and hematopoiesis are unknown. Here, we established tmem106bb-/- knockout (KO) zebrafish and showed that tmem106bb KO reduced the proliferation of HSPCs during definitive hematopoiesis. The differentiation potential of HSPCs to lymphoid lineage was reduced, whereas the myeloid and erythroid differentiation potentials of HPSCs were increased in tmem106bb-/- zebrafish. Similar results were obtained with morpholino knockdown of tmem106bb. Mechanistically, TMEM106B interacted with LAMP2A, the lysosomal associated membrane protein 2A, impaired LAMP2A-Cathepsin A interaction, and enhanced LAMP2A stability; tmem106bb KO or TMEM106B knockdown caused LAMP2A degradation and impairment of chaperone-mediated autophagy (CMA). Knockdown of lamp2a caused similar phenotypes to that in tmem106bb-/- zebrafish, and overexpression of lamp2a rescued the impaired phenotypes of HSPCs in tmem106bb-/- embryos. These results uncover a novel molecular mechanism for the maintenance of HSPC proliferation and differentiation through stabilizing LAMP2A via TMEM106B-LAMP2A interaction.


Subject(s)
Cell Differentiation , Cell Proliferation , Hematopoietic Stem Cells , Lysosomal-Associated Membrane Protein 2 , Membrane Proteins , Zebrafish , Animals , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Lysosomes/metabolism , Humans , Hematopoiesis/physiology
19.
Sci Rep ; 14(1): 19654, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179703

ABSTRACT

Recently, we have reported that extracellular vesicles (EVs) from the bone marrow mesenchymal stromal cells (BM-MSC) of aplastic anemia (AA) patients inhibit hematopoietic stem and progenitor cell (HSPC) proliferative and colony-forming ability and promote apoptosis. One mechanism by which AA BM-MSC EVs might contribute to these altered HSPC functions is through microRNAs (miRNAs) encapsulated in EVs. However, little is known about the role of BM-MSC EVs derived miRNAs in regulating HSPC functions in AA. Therefore, we performed miRNA profiling of EVs from BM-MSC of AA (n = 6) and normal controls (NC) (n = 6) to identify differentially expressed miRNAs. The Integrated DEseq2 analysis revealed 34 significantly altered mature miRNAs, targeting 235 differentially expressed HSPC genes in AA. Hub gene analysis revealed 10 HSPC genes such as IGF-1R, IGF2R, PAK1, PTPN1, etc., which are targeted by EV miRNAs and had an enrichment of chemokine, MAPK, NK cell-mediated cytotoxicity, Rap1, PI3k-Akt, mTOR signalling pathways which are associated with hematopoietic homeostasis. We further showed that miR-139-5p and its target, IGF-1R (hub-gene), might regulate HSPC proliferation and apoptosis, which may serve as potential therapeutic targets in AA. Overall, the study highlights that AA BM-MSC EV miRNAs could contribute to impaired HSPC functions in AA.


Subject(s)
Anemia, Aplastic , Extracellular Vesicles , Gene Expression Profiling , Mesenchymal Stem Cells , MicroRNAs , Anemia, Aplastic/genetics , Anemia, Aplastic/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Humans , Mesenchymal Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Female , Male , Adult , Middle Aged , Hematopoiesis/genetics , Apoptosis/genetics , Bone Marrow Cells/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL