Home Biology The miR-221/222 cluster regulates hematopoietic stem cell quiescence and multipotency by suppressing each Fos/AP-1/IEG pathway activation and stress-like differentiation to granulocytes

The miR-221/222 cluster regulates hematopoietic stem cell quiescence and multipotency by suppressing each Fos/AP-1/IEG pathway activation and stress-like differentiation to granulocytes

0
The miR-221/222 cluster regulates hematopoietic stem cell quiescence and multipotency by suppressing each Fos/AP-1/IEG pathway activation and stress-like differentiation to granulocytes

[ad_1]

Summary

All through life, hematopoietic stem cells (HSCs), residing in bone marrow (BM), repeatedly regenerate erythroid/megakaryocytic, myeloid, and lymphoid cell lineages. This steady-state hematopoiesis from HSC and multipotent progenitors (MPPs) in BM might be perturbed by stress. The molecular controls of how stress can affect hematopoietic output stay poorly understood. MicroRNAs (miRNAs) as posttranscriptional regulators of gene expression have been discovered to regulate varied capabilities in hematopoiesis. We discover that the miR-221/222 cluster, which is expressed in HSC and in MPPs differentiating from them, perturbs steady-state hematopoiesis in methods similar to stress. We evaluate pool sizes and single-cell transcriptomes of HSC and MPPs in unperturbed or stress-perturbed, miR-221/222-proficient or miR-221/222-deficient states. MiR-221/222 deficiency in hematopoietic cells was induced in C57BL/6J mice by conditional vav-cre-mediated deletion of the floxed miR-221/222 gene cluster. Social stress in addition to miR-221/222 deficiency, alone or together, diminished HSC swimming pools 3-fold and elevated MPPs 1.5-fold. It additionally enhanced granulopoisis within the spleen. Moreover, mixed stress and miR-221/222 deficiency elevated the erythroid/myeloid/granulocytic precursor swimming pools in BM. Differential expression analyses of single-cell RNAseq transcriptomes of unperturbed and pressured, proficient HSC and MPPs detected greater than 80 genes, selectively up-regulated in pressured cells, amongst them fast early genes (IEGs). The identical differential single-cell transcriptome analyses of unperturbed, miR-221/222-proficient with poor HSC and MPPs recognized Fos, Jun, JunB, Klf6, Nr4a1, Ier2, Zfp36—all IEGs—in addition to CD74 and Ly6a as potential miRNA targets. Three of them, Klf6, Nr4a1, and Zfp36, have beforehand been discovered to affect myelogranulopoiesis. Along with elevated ranges of Jun, Fos varieties elevated quantities of the heterodimeric activator protein-1 (AP-1), which is thought to regulate the expression of the selectively up-regulated expression of the IEGs. The comparisons of single-cell mRNA-deep sequencing analyses of socially pressured with miR-221/222-deficient HSC establish 5 of the 7 Fos/AP-1-controlled IEGs, Ier2, Jun, Junb, Klf6, and Zfp36, as frequent activators of HSC from quiescence. Mixed with stress, miR-221/222 deficiency enhanced the Fos/AP-1/IEG pathway, prolonged it to MPPs, and elevated the variety of granulocyte precursors in BM, inducing selective up-regulation of genes encoding warmth shock proteins Hspa5 and Hspa8, tubulin-cytoskeleton-organizing proteins Tuba1b, Tubb 4b and 5, and chromatin transforming proteins H3f3b, H2afx, H2afz, and Hmgb2. Up-regulated in HSC, MPP1, and/or MPP2, they seem as potential regulators of stress-induced, miR-221/222-dependent elevated granulocyte differentiation. Lastly, stress by serial transplantations of miR-221/222-deficient HSC selectively exhausted their lymphoid differentiation capacities, whereas retaining their means to residence to BM and to distinguish to granulocytes. Thus, miR-221/222 maintains HSC quiescence and multipotency by suppressing Fos/AP-1/IEG-mediated activation and by suppressing enhanced stress-like differentiation to granulocytes. Since miR-221/222 can be expressed in human HSC, managed induction of miR-221/222 in HSC ought to enhance BM transplantations.

Introduction

All through life, hematopoietic stem cells (HSCs) reside in bone marrow (BM), from the place they proceed to generate, at regular, unperturbed state, all hematopoietic cell lineages [17]. When transplanted, HSCs reconstitute hematopoietic stem and progenitor cell compartments and all hematopoietic cell lineages in regular numbers within the host. At steady-state activation, HSCs enter cell cycle, and, when differentiating, give rise to multipotent progenitors (MPP1-MPP4), which generate frequent lymphoid (CLP) and to frequent myeloid progenitors (CMPs). HSCs are heterogeneous, with multilineage-potential, myeloid-erythroid-biased, myeloid-erythroid-restricted, and differentiation-inactive HSC [3,8,9]. How this heterogeneity is regulated is poorly understood.

Hematopoiesis might be perturbed by completely different types of stress [1,4,1024]. Earlier than “ex vivo/in vitro” preparation of BM cells for FACS analyses, a 20-hour-long transportation of the mice from the breeding facility to the laboratory induces social stress [12]. This social stress is reversible by a 14- to 21-day relaxation interval as it is suggested after housing mice in a brand new setting. “Ex vivo/in vitro” stress might be solely certainly one of a number of types of stress, which affect HSC, when they’re transplanted repeatedly in serial transplantations to review their homing and reconstitution potential [1,4]. We evaluate pool sizes of HSC and of progenitor compartments (MPP1 and a couple of), in addition to single-cell transcriptomic describing their gene expression applications, unperturbed at regular state with these perturbed by both social stress, “in vitro” stress, or transplantation.

MicroRNAs (miRNAs) as posttranscriptional regulators of gene expression have been proven to play a vital position in hematopoiesis, modulating single or a number of genes in a number of mobile capabilities [2530]. Particular ablation of Dicer in HSCs, thus full deletion of mature miRNAs, results in elevated apoptosis in HSCs and impacts their self-renewal capability [3032]. A number of miRNAs have been recognized, which affect the laws of those completely different states of HSC [31,33] by the mTOR pathway [34,35].

The cluster of miR-221 and miR-222 was discovered to be expressed in early hematopoietic progenitors [3537]. Aside from a earlier report that indicated a possible involvement of the miR-221/222 cluster in early erythropoiesis by regulating equipment protein expression, the position of this microRNA cluster in early hematopoiesis remained unknown [38]. Moreover, we’ve beforehand described the influences of miR-221 on preBI-cell homing to BM [39]. MiR-221 overexpression confers the power to those cells to residence to, and turn out to be resident in particular, subosteal areas within the BM after transplantation. That is completed by activation of the PI3K signaling community in response to BM area of interest elements just like the chemokine CXCL12. This activation induces integrin VLA-4 to vary to its excessive affinity binding conformation, which permits elevated adhesion and residence in environments of BM expressing the cell adhesion molecule VCAM1 [39]. Since HSCs have the pure means to residence to and engraft in BM, we reasoned that miR-221/222 would possibly affect HSC swimming pools in BM by regulating their engraftment as in preB cells. As we present right here, this speculation isn’t tenable for HSC.

Right here, we present that every one HSC and MPP categorical the miRNA cluster. We then generate mice, during which HSC and their subsequent lineages turn out to be targetable for deletion of the floxed miR-221/222 cluster, utilizing Vav-promotor-induced expression of iCre [9,40]. Single-cell expression analyses present that every one HSC and MPPs have their miR-221/222 cluster deleted.

Deletion of the miR-221/222 cluster ends in diminished numbers of HSC throughout steady-state hematopoiesis, accompanied by elevated numbers of MPPs (MPP1-MPP4) just like the impact of social stress perturbation in miR-221/222-proficien mice. Importantly, the variety of granulocytes in spleen is elevated by the miR-221/222 deficiency.

MiR-221/222 deficiency mixed with social stress additional enhances granulopoiesis by rising numbers of granulocyte progenitors in BM.

With a view to perceive the molecular mechanisms of stress perturbation and/or miR-221/222 deficiency, we generate a single-cell transcriptional panorama of hematopoietic progenitors in BM.

Single-cell transcriptomes of mixed socially pressured, miR-221/222-deficient HSC and MPPs recognized selectively up-regulated expression of genes encoding warmth shock proteins, tubulin-cytoskeleton-organizing proteins, and chromatin transforming proteins. Lastly, mixed stress induced by serial transplantations and miR-221/222 deficiency exhausts lymphoid differentiation, however not BM-homing and granulocyte differentiation capacities of HSC.

Outcomes

Managed perturbation of steady-state hematopoiesis by “in vivo” social stress reduces HSC swimming pools and will increase MPPs in BM

Social stress imposed for lower than 1 day permitted us to measure and to check the actions of social stress on pool sizes of miR-221/222-proficient BM cells. We discovered that inside 24 hours, social stress diminished the HSC pool in BM 2-fold and elevated MPP2 two-and-a-half-fold and MPP4 2.2-fold (Fig 1A; for nomenclature, see Wilson and colleagues and Cabezas-Wallscheid and colleagues [5,7]). We additionally analyzed if the impact of transportation-induced social stress on stem cell populations of BM is reverted to regular state by time. For this, we analyzed the numbers of cells 3 days, 7 days, and 21 days after transportation. We discovered that, with important fluctuations, the variety of HSC, MPP1, MPP2, MPP3, and MPP4 turns into indistinguishable from the unperturbed pool of cells 3 weeks after social stress induction (Fig 1A). We didn’t detect important variations at any time factors after social stress perturbation in numbers of frequent lymphoid (CLP), megakaryocyte-erythroid (MEP), frequent myeloid (CMP), and granulocyte-myelocyte (GMP) progenitors in BM of miR-221/222-proficient mice (S1A Fig).

thumbnail

Fig 1. The impact of in vivo social stress perturbation on hematopoietic cells in BM.

(A) The evaluation of stream cytometry measurements on BM HSCs MPP1, 2, 3, and 4 cells with out (unperturbed) and 1 day, 3 days, 7 days, or 21 days after short-term in vivo social stress perturbation. Single-cell suspensions from 2 tibia and a couple of femurs/mice of unperturbed (Unpert./open circle) or in vivo social stress–perturbed (open squares at completely different time factors) C57B6/J mice had been stained for stream cytometry, measured, and the overall numbers of cells/mice had been plotted. Purple strains point out the imply values. One-way ANOVA with Tukey publish normalization check was used to guage statistical significance (*, **, and *** point out p < 0.05, p < 0.01, and p < 0.001, respectively. Knowledge can be found in S1 Knowledge and on FlowRepository.org by FR-FCM-Z6PS accession quantity). (B) Genes with larger expression after short-term in vivo social stress perturbation (purple dots and gene symbols are chosen genes) in HSC are introduced by comparative differential expression evaluation of unperturbed versus perturbed cells. Differentially expressed genes are above the importance restrict. The log2 fold-change expression values had been plotted towards the numbers of unperturbed cells categorical the gene. (C) The transcriptomes of single HSC, MPP1, and MPP2 cells of unperturbed or social stress–perturbed mice had been analyzed by droplet-based scRNA sequencing. The samples had been collected most 1 day after social stress induction. The clustering of early (E) hematopoietic compartment are referred as E-Clusters 0–6. (D) The frequency distribution (purple to blue) of unperturbed and social stress–perturbed HSC, MPP1, and MPP2 cells had been projected to UMAPs with a purpose to present transcriptional program modifications induced by social stress. (E) Expression of gene-set modules (“Non-proliferating,” “Cell cycle,” and “IEG”) and matched frequencies of cells expressing the gene-set within the completely different clusters are introduced as Bubble-plots. (F) The distribution of sorted unperturbed (unpert.) and short-time in vivo social stress–perturbed (pert.) HSC, MPP1, and MPP2 cells is proven within the aggregated E-clusters. The frequency of a given cell sort in E-clusters is coloured from highest to lowest respective purple to blue. The numerical information for Fig 1B–1F might be present in S2 Knowledge. BM, bone marrow; HSC, hematopoietic stem cell; IEG, fast early gene; MPP, multipotent progenitor; UMAP, Uniform Manifold Approximation and Projection for Dimension Discount.


https://doi.org/10.1371/journal.pbio.3002015.g001

Perturbation by “in vivo” social stress induces elevated transcription of chosen genes, amongst them IEG in HSC, MPP1, and MPP2 cells

Subsequent, we targeted our transcriptome analyses on the earliest progenitors HSC, MPP1, and MPP2 to watch early modifications in gene expression, imposed by the change from the regular state to the social stress–perturbed state of HSC, MPP1, and MPP2 cells. All sequencing information had been analyzed at the same depth, displaying comparable variety of genes and mRNAs per cell, permitting quantitative differential gene expression analyses.

We outlined differentially expressed genes as expressed by (i) greater than 30% of all unperturbed cells, (ii) with a considerably larger log2 fold-change between unperturbed and “in vivo” social stress–perturbed cells, when adjusted to the 99% prediction band of a linear regression mannequin. Genes above the noise degree (red-dashed strains in Figs 1B and S2) are thought-about as considerably larger expressed in social stress–perturbed cells.

Comparability of gene expression ranges in unperturbed and in socially pressured, perturbed HSC (Fig 1B), MPP1, or MPP2 (S2 Fig) 1 day after stress induction detected a perturbation-dependent normal improve and an much more pronounced improve within the ranges of transcription of IEGs beside a small set of roughly 90 genes in these cells (Figs 1B and S2). This implies that perturbation of hematopoiesis by “in vivo” stress would possibly use IEG-controlled activation.

Comparability of entire transcriptomes of unperturbed with socially perturbed HSC, MPP1, and MPP2 detects variations in clusters of gene expressions

In major cluster evaluation of the transcriptomes of unperturbed and socially perturbed HSC, MPP1, and MPP2, 7 E-clusters (E for early hematopoiesis) had been recognized (Fig 1C), and the cell frequency distributions had been projected in UMAPs (Fig 1D). E-0 cluster represents nonproliferating cells (primarily from HSC), E-1 activated towards cell cycle, and E-2 activated towards IEG expression (Fig 1E). By location on the UMAP, E-2 isn’t directed towards proliferating clusters, however its place might point out another activation. E-3-6 comprise MPP1 and MPP2 expressing G1/S and G2/M cell cycle genes (Figs 1E and S3A and S2 Knowledge).

In contrast with unperturbed cells, social stress–perturbed cells in E-clusters had been 14% much less in nonproliferating E-0, 4% extra in proliferation-activated E-1, 9% extra in E-3, and 13% extra in E-2 (Fig 1F).

The E-2 cluster contained IEG signatures (Fig 1E). Since its place inside the UMAP panorama of clusters of transcriptionally relates cells is outdoors the G1/S/G2 cell cycle–lively clusters E1 to E6, they might be in another way activated. (Fig 1D and 1E). These outcomes present that early progenitors are activated from quiescence by the stimulatory exercise of short-term perturbation.

The miR-221/222 gene cluster is expressed and might be deleted in all HSC and MPPs

The miR-221/222 gene cluster is situated on the X chromosome and is expressed in HSC, in activated multipotent progenitors MPP1 and in proliferating MPP2, in myeloid-lymphoid-directed MPP3 and MPP4, and in CLP, granulocytes, and myeloid cells. We discovered that miR-221/222 cluster is turned off in BM preB cells, in thymocytes, and in peripheral blood derived, largely nonactivated pure killer (NK) cells T and B lymphocytes (S4A Fig). These outcomes assist earlier findings that resting cells don’t categorical the miRNA household, however Th17 peripheral T cell subpopulations improve miR-221/222 expression upon activation [41]. It has been additionally described that miR-221/222 elevated expression performs an necessary position in activated B cell Ig class change in germinal middle reactions [42].

We decided what number of single HSC, MPP1, and MPP2 categorical what number of miR-221 and miR-222 molecules per cell, by a normal curve-based, multiplexed stem-loop RT-TaqMan qPCR assay. We discovered that each examined HSC, MPP1, and MPP2 cell expressed between 7,000 and eight,000 copies of miR-221. Expression of miR-222, once more examined in each cell, was decrease and extra variable between 1,000 and 5,000 copies per cell (Fig 2A). The extent of miR-221 expression strongly correlated with miR-222 expression (Fig 2B).

thumbnail

Fig 2. Expression evaluation of miR-221/222 in single cells and the impact of miRNA deficiency on hematopoietic stem cell populations in BM.

(A) Copy numbers of miR-221 (closed sq.) and miR-222 (open sq.) measured in single HSC, MPP1, and MPP2 of miRNA-proficient and in swimming pools of HSC+MPP1 single cells of miRNA-deficient mice (ncell = 36 cells from 3 mice, nNTC = 12 wells). The best worth of NTC is marked as straight line, indicating the detection restrict. (B) Correlation evaluation of miR-221 and miR-222 copy numbers in proficient HSC, MPP1, and MPP2 cells (C, D) The evaluation of stream cytometry measurements on BM and spleen derived cells. (C) Single-cell suspensions from 2 tibia and a couple of femurs/mice or (D) from spleens of miR-221/222-proficient (open sq.) or miR-221/222-deficient (closed sq.) mice had been stained for stream cytometry, measured, and the overall numbers of cells/mice had been plotted. Purple strains point out the imply values. Pupil t check was used to guage statistical significance (* or ** point out p < 0.05 or p < 0.01, respectively. The numerical information for the graphs might be present in S1 Knowledge and on FlowRepository.org by FR-FCM-Z6PS accession quantity). BM, bone marrow; HSC, hematopoietic stem cell; miRNA, microRNA; MPP, multipotent progenitor; NTC, no-template management.


https://doi.org/10.1371/journal.pbio.3002015.g002

To check capabilities of miR-221/222 throughout hematopoiesis, we generated mice, during which the one miR-221/222 cluster on the X chromosome in male mice might be deleted in all hematopoietic cells. Vavi-cre, lively in all hematopoietic cells together with HSCs, is often used to drive Cre recombinase expression in hematopoietic cells [4,40]. Thus, Vav-Cre mice had been crossed with miR-221/222fl/fl mice to generate male F1 progeny, the place the miR-221/222 cluster was anticipated to be deleted on the one X chromosome. All mouse strains had been backcrossed on C57BL/6J for a minimum of 8 generations to establish genetic homogeneity, aside from the miR-221/222 cluster in all offsprings. Bulk-sorted BM-lineage (lin)c-kit+Sca1+ (LSK), and MPP cells (LSK CD150CD48+) (S4B Fig), and single LSK CD150+CD48 (pool of HSC and MPP1) cells of miR-221/222fl/y-Tg(Vav1-icre) mice had been examined for quantitative miR-221/222 expression on the single-cell degree. Certainly, our analyses present excessive effectivity of deletion of the miR-221/222 cluster in all examined cell populations. (Figs 2A and S4B).

MiR-221/222 deficiency decreases HSC and will increase MPP in unperturbed hematopoiesis

In BM of miR-221/222-deficient mice at regular state of unperturbed hematopoiesis, we discovered numbers of HSC diminished roughly 3-fold, whereas MPP2 and MPP3 had been considerably elevated. (Fig 2C). The extent of those modifications within the numbers of HSC, MPP1, and MPP2 are just like these seen in socially perturbed HSC, MPP1, and MPP2. This might recommend that miR-221/222 deficiency and social stress would possibly use, a minimum of in components, the identical mechanisms to manage pool sizes of early progenitors.

Numbers of MPP1, MPP4 (Fig 2C), CLP, CMP (S1B Fig) and MEPs and GMPs (Fig 5B) and peripheral CD4+, CD8+ T cells, and CD19+ B cells in spleen weren’t modified (S1C Fig). Apparently, in spleen, the numbers of granulocytes had been 6-fold elevated in miR-221/222-deficient mice (Fig 2D; information additionally on S1C Fig). These outcomes recommend that expression of miR-221/222 maintains pool sizes of HSC and MPPs and prevents an elevated granulopoiesis.

MiR-221/222 targets Fos to guard the dimensions of the unperturbed HSC pool

Subsequent, we in contrast transcriptomes of poor with proficient cells to detect microRNA-sensitive genes. We outlined these genes as expressed by (i) greater than 30% of all miR-221/222-proficien cells, (ii) with a considerably larger log2 fold-change between proficient and miR-221/222-deficient cells, when adjusted to the 99% prediction band of a linear regression mannequin. Genes above the noise degree (red-dashed strains in Fig 3A, 3E and 3F) are thought-about as considerably expressed in miR-221/222-deficient cells.

thumbnail

Fig 3. miR-221/222-sensitive genes in unperturbed HSC, MPP1, and MPP2 cells.

(A) Comparative differential expression evaluation between miR-221/222-deficient and proficient HSC, (E) MPP1, and (F) MPP2 cells. The log2 fold-change expression values are plotted towards the numbers of proficient cells expressing the gene. After calculating the linear regression curve, the 99% CI band (dashed purple strains) decided the nondifferentially (grey dots) and differentially (purple dots and gene symbols) expressed genes. Considerably larger expressed genes in miR-221/222-deficient cells are above the 99% CI restrict and are expressed by greater than 30% of all cells (inexperienced line). These genes are known as “differentially expressed genes.” (B) Single-cell expression of Fos in HSC, MPP1, and MPP2 cells. The distinction within the imply expression degree of the indicated populations between proficient (open half violin) and poor (crammed half violin) cells are introduced. (C) The RQ of Fos mRNA in bulk sorted miR-221/222-proficient (open bars) and miR-221/222-deficient (crammed bar) HSC and MPP1 cells measured by RT-qPCR. RQ was calculated on the idea of Hprt expression in miR-221/222-proficient HSC. (D) The differential expression of AP-1 elements Fos and Jun introduced on UMAP of unperturbed miR-221/222-proficient and miR-221/222-deficient HSC, MPP1, and MPP2 cells. The numerical information for Fig 3A, 3B, and 3D–3F might be present in S2 Knowledge, and the numerical information for Fig 3C might be present in S1 Knowledge. AP-1, activator protein-1; CI, confidence interval; HSC, hematopoietic stem cell; MPP, multipotent progenitor; RQ, relative amount; RT-qPCR, quantitative reverse transcription PCR; UMAP, Uniform Manifold Approximation and Projection for Dimension Discount.


https://doi.org/10.1371/journal.pbio.3002015.g003

At regular state of unperturbed hematopoiesis, we detected 9 such differentially expressed genes (Fig 3A and 3B). Amongst them, Fos was the one miR-221/222-seed-sequence-containing goal gene (DIANA-TarBase v8 [43], TargetScan Launch 8.0 [44], miRDB [45]), which has been experimentally validated by Errico and colleagues [46]. We validated this elevated Fos expression with qPCR analyses (Fig 3C), during which Fos was discovered to be expressed 8.5-fold larger in poor HSC. Six of the opposite 8 genes (Jun, JunB, Nr4a1, Ier2, Zfp36, and Klf6) are like Fos IEG [47,48] (Fig 3A and 3D).

Subsequent, we carried out the identical analyses with MPP1 and MPP2. In unperturbed MPP1, Fos, JunB, Nr4a1, Cd74, Ly6a, Zfp36, and Klf6 had been not detectable, and Rgs1 appeared (Fig 3D and 3E). In MPP2, no differentially expressed genes remained detectable (Fig 3F). This implies that, in unperturbed hematopoiesis, up-regulated mRNA expression of Fos, Jun, and 5 different IEG transcription elements act primarily in HSC, inflicting a lower of HSC and a rise of MPPs and of granulocytes.

We now have additionally analyzed with gene set enrichment evaluation (GSEA; [49,50]) if the expression of predicted miR-221/222 goal genes (Fig 4A) have differential distribution in miR-221/222-deficient HSC, MPP1, and MPP2 cells based on E-clusters. These analyses present that the goal gene expression is sort of equally distributed with out important accumulation in any E-cluster between HSC, MPP1, and MPP2 cells (Fig 4B and S2 Knowledge). We additionally couldn’t detect important distinction within the cumulative expression between miR 221/222-proficient and miR 221/222-deficient cells as proven in Fig 4C and S2 Knowledge. Moreover, we plotted the differential expression of every of the expected goal genes towards the frequency of cells expressing the gene. We discovered 2-to-3 genes having marginal (roughly 5%) larger expression in a minimum of 10% of poor cells (Fig 4D–4F). Value noting that 2-to-3 genes had been additionally discovered to be larger expressed on the marginal degree (roughly 5%) in miR-221/222-proficient cells, indicating a stochastic differential expression worth across the 5% boundary. We discovered solely Fos as experimentally validated direct goal expressed roughly 20% larger in additional than 60% of miR 221/222-deficient HSC and MPP1 cells.

thumbnail

Fig 4. GSEA and differential gene expression evaluation of predicted miR 221/222 goal genes in scRNA-seq information of miR-221/222-proficient and miR-221/222-deficient HSC, MPP1, and MPP2 cells.

(A) Collection of 310 predicted miR-221/222 goal genes recognized by a minimum of 2 of three bioinformatical miRNA goal prediction instruments (TarBase v8, TargetScan Launch 8.0 and miRDB). (B) GSEA was carried out for every cell based mostly on the distinction to the imply of log normalized expression values of all cells within the analyzed set as preranked checklist and 1,000 randomizations. Vital up- or down-regulation was outlined by an FDR ≤ 0.50 and normalized p-value < 0.05. For visualization, NES for important cells had been plotted. The GSEA was carried out for indicated cells utilizing the 310 predicted miR-221/222 goal genes. (C) Violin plots present the imply (purple bars) and median (black bars) variations of the NES of the GSEA in miR-221/222-proficeient and miR-221/222-deficient HSC, MPP1, and MPP2 cells. Vital variations between the NES values had been calculated by Mann–Whitney U check. (D-F) Differential expression analyses of predicted miR-221/222 goal gene expressions in miR-221/222-proficient and miR-221/222-deficient HSC (D), MPP1 (E), and MPP2 (F). The purple dashed line signifies the 5% differential expression band. The numerical information for Fig 4A–4F might be present in S2 Knowledge. FDR, false uncover charge; GSEA, gene set enrichment evaluation; HSC, hematopoietic stem cell; MPP, multipotent progenitor; NES, normalized expression rating.


https://doi.org/10.1371/journal.pbio.3002015.g004

Shared IEG expression up-regulated by social stress perturbation and miR-221/222 deficiency

Subsequent, we in contrast the differentially up-regulated genes in transcriptomes of social stress–perturbed and of experimentally unperturbed, miR-221/222 deficiency–influenced HSC, MPP1, and MPP2 cells. Six IEGs—Fos, Jun, JunB, Ier2, Klf6, and Zfp36–had been discovered up-regulated by each stressors (Fig 5A). This implies that HSC and MPPs use IEG responses to regulate pool sizes of HSC and their granulocyte biased differentiation. The outcomes of our experiments point out that this pathway is managed by the miR-221/222 gene cluster.

thumbnail

Fig 5. Shared gene expression program in social stress–perturbed and miR-221/222 deficiency–induced HSC and MPP1 cells.

(A) The in vivo social stress–perturbed and miR-221/222 deficiency–induced genes had been decided by differential gene expression analyses of scRNA-seq information and the plotted. The world-proportional Venn diagram exhibits the commonality between short-term social stress perturbation induced and miR-221/222 deficiency–delicate genes. The complete gene checklist is out there from S2 Knowledge. (B) Evaluation of stream cytometry measurements on unperturbed and social stress–perturbed MEPs and GMPs. Single-cell suspensions of tibia and femurs of unperturbed miR-221/222-proficient (open squares) and miR-221/222-deficient (closed squares) mice or perturbed miR-221/222-proficient (open triangles) or miR-221/222-deficient (closed triangle) mice had been ready, analyzed with stream cytometry, and the numbers of cells had been plotted. Purple strains point out the imply values. One-way ANOVA with Tukey posttest was used to guage statistical significance (*, **, and *** point out p < 0.05, p < 0.01, and p < 0.001, respectively. Knowledge can be found in S1 Knowledge, in S2 Knowledge, and on FlowRepository.org by FR-FCM-Z6PS accession quantity). GMP, granulocyte-myelocyte progenitor; HSC, hematopoietic stem cell; MEP, megakaryocyte-erythroid progenitor; MPP, multipotent progenitor.


https://doi.org/10.1371/journal.pbio.3002015.g005

Managed perturbation by social stress of miR-221/222-deficient BM results in will increase in numbers of MEP and GMP granulocyte progenitors

In unperturbed, miR-221/222-deficient mice elevated numbers of granulocytes had been noticed as a doable involvement of this miRNA cluster in enhanced granulopoiesis (Fig 2D). In assist of this notion, brief social stress (20 hours) was ample to induce will increase within the numbers of MEP and GMP (in common 4-fold every) in miR-221/222-deficient, however not in miR-221/222-proficient BM (Fig 5B). The variety of CLP and CMP was not completely different (S1A and S1B Fig). These fast modifications in granulocyte progenitor compartment sizes recommend that miR-221/222-deficient, pressured HSC and MPPs want solely a brief interval of perturbation, during which proliferation and differentiation improve granulocyte-biased, however not lymphoid-directed hematopoiesis, detectable in elevated numbers of MEP and GMP. In contrast, stress-induced modifications in HSC and MPP numbers should not additional altered by miRNA deficiency. Due most likely to the brief interval of activation, the numbers of mature granulocytes within the periphery should not (but) additional elevated (S1B Fig). We conclude from these outcomes that miR-221/222 safeguards myeloid-lymphoid multipotency of HSC and MPP additionally in pressured hematopoiesis.

Transcriptional landscapes of hematopoietic progenitor cells detect miR-221/222 deficiency–induced granulopoiesis

With a view to assemble complete transcriptional landscapes of hematopoietic progenitors in BM at unperturbed, steady-state hematopoiesis, with trajectories of gene expressions throughout growth to completely different lymphoid, myeloid, and erythroid cell lineages and to detect doable modifications in granulocyte-directed trajectories, we performed separate transcriptome analyses for HSC, MPP1, and MPP2, together with additionally extra mature BM progenitors, i.e., MPP3, MPP4, CLP, and linc-kit+Sca1 or linc-kitSca1 cells from miR-221/222-proficient and miR-221/222-deficient BM. Cells had been clustered (T-clusters for “complete”) based on their transcriptomes by Uniform Manifold Approximation and Projection for Dimension Discount (UMAP) [51] (Fig 6A).

thumbnail

Fig 6. Single-cell transcriptome evaluation of unperturbed miR-221/222-proficient and miR-221/222-deficient lineage-negative BM compartment.

(A) Cluster evaluation on the aggregated UMAP plots of miR-221/222-proficient and miR-221/222-deficient HSC, MPP1, MPP2, MPP3-4 pool, CLP, linckit+Sca1, and linckitSca1 populations (complete, T-clusters) after single-cell transcriptome sequencing. Proficient and poor cells are individually plotted. (B) The bubble-plots exhibits the expression of gene-set modules (z-score) and matched frequencies of cells (bubble dimension) expressing the gene-set within the completely different T-clusters. miR-221/222-proficient and miR-221/222-deficient samples had been individually plotted. (C, D) Main hematopoietic differentiation pathways are introduced on the UMAP as attribute gene-set modules (S2 Knowledge) by the Log2 abstract expression of the module genes. (C) Cells with attribute expression sample for Mye (to Myeloid), T (to T cells), B (to B cells), Ery (to erythrocytes), Thr (to thrombocytes) are proven on a composite image for proficient and poor cells and (D) for Ba/Ma (to basophil/mast cells) and Neu/Gran. (to neutrophils/granulocytes). (E) Frequencies of sorted miR-221/222-proficient and miR-221/222-deficient HSC, MPP1, MPP2, MPP3-4 pool, CLP, linckit+Sca1, and linckitSca1 cells in several T-clusters. The frequency of a given cell sort in T-clusters is coloured from highest to lowest respective purple to blue. (F) Relative frequencies of cells in several T-clusters are introduced on Log10 scale (frequency of poor cells relative to the frequency of proficient cells). The numerical information might be present in S2 Knowledge. BM, bone marrow; CLP, frequent lymphoid progenitor; HSC, hematopoietic stem cell; MPP, multipotent progenitor; UMAP, Uniform Manifold Approximation and Projection for Dimension Discount.


https://doi.org/10.1371/journal.pbio.3002015.g006

We chosen units of genes to characterize expression applications for HSC, cell -cycle, erythroid, megakaryocytic, lymphoid, myeloid, and granulocyte differentiation and assigned these T-clusters (Figs 6B–6E and S3B and S2 Knowledge). These analyses are in settlement with earlier publications of different laboratories [18,52].

Solely 2 T-clusters had been completely different in proficient and poor cells. T-7 (basophil/mast cell) contained −1.9-fold much less, T-11 (granulocytes) 2.9-fold extra cells in miR-221/222-deficient mice (Fig 6E and 6F). Actually, the granulocyte-related genes had been solely current in T-11 of miR-221/222-deficient, however not of proficient granulocytes (Fig 6B and 6D, decrease left- and right-hand corners).

These outcomes assist our findings (Fig 5B) of elevated numbers of granulocyte precursors in BM and granulocytes within the spleen miR-221/222-deficient mice. They recommend that steady-state hematopoiesis of miR-221/222-deficient BM progenitor cells improve granulopoiesis even in an experimentally unperturbed state.

In perturbed HSC, MPP1, and MPP2 cells, miR-221/222 deficiency selectively will increase transcription of genes encoding warmth shock proteins, G protein–mediated tubulin, and chromatin transforming actions

Lastly, we in contrast transcriptomes of miR-221/222-deficient with proficient cells to detect microRNA-sensitive genes after “in vivo” social stress perturbation. We outlined differentially expressed genes as expressed by (i) greater than 30% of all in vivo social stress–perturbed miR-221/222-proficient cells, (ii) with a considerably larger log2 fold-change between in vivo social stress–perturbed miR-221/222-proficient and miR-221/222-deficient cells, when adjusted to the 99% prediction band of a linear regression mannequin. Genes above the noise degree (red-dashed strains in Fig 7A–7C) are thought-about as considerably expressed in in vivo social stress–perturbed miR-221/222-deficient cells.

thumbnail

Fig 7. Evaluation of in vivo social stress perturbation–delicate genes in miR-221/222-deficient HSC, MPP1, and MPP2 cells.

Genes with larger expression after short-term perturbation (purple dots and gene symbols are chosen genes) in miR-221/222-deficient (A) HSC, (B) MPP1, and (C) MPP2 are introduced by comparative differential expression evaluation of unperturbed versus perturbed cells. Differentially expressed genes are above the importance restrict. The log2 fold-change expression values had been plotted towards the numbers of unperturbed cells categorical the gene. The numerical information might be present in S2 Knowledge.


https://doi.org/10.1371/journal.pbio.3002015.g007

Beside up-regulated IEG expression, social stress induction mixed with miR-221/222 deficiency selectively up-regulated 3 teams of functionally related genes in HSC, MPP1, and/or MPP2 (Fig 7A–7C):

  1. Warmth shock protein (HSP70) genes Hspa5 and Hspa8, with potentialities to activate the unfolded protein response (UPR) [53].
  2. G protein signaling (Rgs1) and its tubulin elements (Tubb1b, 4b, and 5) in cytoskeleton and microtubule meeting, spindle formation, and mitotic cell cycle management [54]. Past the recognized actions of Rgs1 in hypoxia [55] and in SDF-1/CXCR4-induced migration of HSC [56], these genes are anticipated to contribute to elevated erythroid-myeloid differentiation.
  3. Replication-independent, histone-associated chromatin remodelers (H3f3b, Hmgb2, H2afx, H2afz). Since H3f3b is thought to induce erythroid differentiation [57], miR-221/222-dependent up-regulation of H3f3b would favor erythroid-myeloid progenitor activation, as seen in our analyses.

We anticipate that the mixed actions of all of those up-regulated genes may contribute to the activation of HSC from quiescence and to induce selectively elevated granulopoiesis [52,5861].

miR-221/222-deficient HSC fail to reconstitute recipients in serial transplantations

In transplantations with the goal to reconstitute the hematopoietic cells of a lethally irradiated host, CD34 HSCs have been discovered to comprise long-term, totally repopulating HSC, whereas CD34+ MPP1 repopulate all lineages however have diminished capacities to reconstitute long-term repopulating HSC and early progenitors [1,4,5]. Serial transplantations of HSC might be anticipated to impose “in vitro” stress on HSC throughout their preparation for transplantations.

With a view to check repopulation capacities of miR-221/222-proficient and of miR-221/222-deficient HSC, we transplanted 100 donor CD45.2+ both proficient or poor HSC into lethally irradiated CD45.1+ mice, along with 106 non-irradiated CD45.1+ BM provider cells recognized to comprise 100 proficient HSCs—situations which might be anticipated to result in 50% CD45.1–50% CD45.2 chimerism. In serial transplantations, we then sorted CD45.2+ HSC from the transplanted mice and re-transplanted 100 HSCs underneath the identical situations.

In blood, the chimerism in complete CD45+ hematopoietic cells was discovered to be shut to those anticipated values after the primary (roughly 40%) and second (roughly 55%) transplantations of miR-221/222-proficient HSC (Fig 8A). With miR-221/222-deficient HSC, this chimerism was considerably decrease 4 months after the primary (roughly 24%) and far decrease 4 months after the second (2,5%) transplantations, suggesting that miR-221/222 deficiency exhausts the capability of HSC for hematopoietic reconstitution.

thumbnail

Fig 8. miR-221/222 cluster safeguards the multipotency of HSC.

(A) Proportion of donor-derived blood cells after the primary (higher) and second (decrease) transplantation of 100 miR-221/222-proficient (open squares) and miR-221/222-deficient (closed squares) HSC. (B) Frequencies of B cells, CD4+/CD8+ T cells, CD11b+Gr1+ neutrophils/granulocytes (Neu/Gran.), and CD11b+Gr1 monocytes/macrophages/dendritic cells (Mo/MF/DC) of donor-derived blood cells after the primary (higher line) or second (decrease line) transplantation. (C) Numbers of donor-derived HSC, MPP1, and MPP2 cells 16 weeks after the primary (higher line) or the second (decrease line) miR-221/222-proficient (open squares) and miR-221/222-deficient (closed squares) HSC transplantation. Purple strains point out the imply values. (A-C) Knowledge from 5 miR-221/222-proficient and from 6 poor mice are introduced as (A, B) imply values or (C) individually. Pupil t check was used to calculate important variations (*, **, and *** point out p < 0.05, p < 0.01, and p < 0.001, respectively. The numerical information for Fig 8A–8C might be present in S1 Knowledge and on FlowRepository.org by FR-FCM-Z6PS accession quantity. (D) Density plots of pooled miR-221/222-proficient and miR-221/222-deficient HSC, MPP1, and MPP2 cells after serial transplantation are visualized on the aggregated UMAP and (E) the distribution of cells within the completely different E-clusters (see clusters in Fig 1D). The frequency of a given cell sort in E-clusters is coloured from highest to lowest respective purple to blue. The numerical information for Fig 8D and 8E might be present in S2 Knowledge. HSC, hematopoietic stem cell; MPP, multipotent progenitor; UMAP, Uniform Manifold Approximation and Projection for Dimension Discount.


https://doi.org/10.1371/journal.pbio.3002015.g008

Regardless of this diminished peripheral chimerism in poor versus proficient T (15% versus 35%) and B cells (10% versus 30%), no variations in reconstitution had been noticed between proficient and poor HSCs in BM 4 months after the second transplantation. Moreover, secondary recipients of proficient HSC had 30% donor-derived myeloid cells and granulocytes, whereas recipients of poor HSC contained 65%. This implies that twice-transplanted miR-221/222-deficient HSC continued to develop granulocytes however had turn out to be poor of their capacities to develop lymphoid cells (Fig 8B).

In BM, serial transplantations of proficient and poor HSC generated completely different sizes of repopulated HSC and MPP2 swimming pools (Fig 8C). We discovered an equal chimerism of proficient in addition to poor CD45.2+ HSC after the primary transplantation, indicating comparable, ample homing to the BM of each sorts of HSC. 4 months after secondary transplantation, proficient HSC established regular HSC ranges. Surprisingly, ranges of twice transplanted poor HSC ranges had been even 2- to 3-fold larger (Fig 8C). Numbers of MPP1 weren’t completely different, whereas MPP2 had been diminished already after the primary, and much more after the second transplantation of poor HSC (Fig 8C). These information point out that twice transplanted miR-221/222-deficient HSC retain their capability to residence to and repopulate their niches in BM however lose components of their capacities of hematopoietic differentiation. Thus, the repeated “in vitro” stress throughout transplantation would possibly favor the preservation, and even enhancement of granulocyte differentiation capacities of miR-221/222-deficient HSC, nevertheless it additionally extinguishes their lymphoid differentiation capabilities.

Serial transplantations of miR-221/222-deficient HSC accumulate cell cycle–inactive HSC and deplete proliferating MPPs

Lastly, we additionally in contrast twice serially transplanted miR-221/222-deficient HSC and MPPs, which had misplaced their multipotency however had retained their BM homing capability (Fig 8A–8C) with proficient, twice-transplanted progenitors. miR-221/222-deficient cells had elevated numbers of E-0 (nonproliferating HSC) however decreased numbers of E-4-6 (cell cycle–lively) (Fig 8D and 8E). This implies that serial transplantation of miR-221/222-deficient HSC accumulates proliferation-inactive HSC and depletes cell cycle–lively MPPs. These poor HSC are harking back to differentiation-inactive HSC clones described by Pei and colleagues [8]. Once more, we conclude from these outcomes that miR-221/222 safeguards the multipotency of HSC.

Dialogue

From the time, when BM develops and is populated in particular niches by long-lived HSC, steady-state hematopoiesis replenishes most central and peripheral hematopoietic cell compartments all through life, with half-lives between a couple of days and some weeks. Most HSC are generated as soon as throughout embryonic growth of the bone and its marrow to stay long-lived, quiescent cells all through life [19]. A smaller a part of these HSCs and extra differentiated progenitors function life-long sources for this steady regeneration. In mouse BM, a couple of thousand LSK CD150+ CD48 CD34 HSC have the capability to outlive as quiescent cells with out dividing a lot for years, even for all times. When transplanted into recipient mice, a single HSC can repopulate the HSC compartment, all progenitor compartments, and all of the mature hematopoietic lineages in regular numbers. Therefore, HSC can discover their niches in BM, can fill these niches by symmetric cell divisions, and might provoke multilineage differentiation to mature erythroid, megakaryocytic, myeloid, and lymphoid cells [19]. Our findings that serial transplantations of pressured, miR-221/222-deficient HSC don’t alter this repopulation capability, however have an effect on hematopoietic multipotency, is perhaps a helpful system to review mechanisms controlling the homing and residence of HSC.

Perturbations of hematopoiesis by bacterial [15] or viral infections [13], by social stress [12], or by transplantation [1,4], mediated by interferon-α [18] or interferon-γ [19], by poly-I:C dsRNA [15], or by G-CSF [20] have all been discovered to affect on steady-state multilineage hematopoiesis of HSC and favor differentiation-restricted HSC, main, e.g., to emergency granulopoiesis [21]. Such restrictions have been seen to turn out to be extra distinguished with growing old [22,24]. Repeated, long-term perturbations of HSC throughout life, e.g., by infections [13], by repeated social stress [12], or throughout serial BM transplantations, as performed in our experiments, all can contribute to HSC growing old [33]. Our findings that social stress favors another activation of HSC to elevated granulopoiesis, that features IEG activation, and that miR-221/222-deficiency prompts additional gene expressions would possibly supply new methods to know the selection of HSC to be activated both to steady-state multilineage differentiation or to stress-induced myelopoiesis and granulopoiesis.

Fos has been validated as direct goal gene in cutaneous melanoma cells [46]. The very low numbers of HSC in a mouse, and the dearth of tissue tradition situations to create bigger numbers of cells, haven’t allowed us to validate Fos as miR-221/222-target in HSC. Bioinformatical instruments predict a minimum of 300 genes expressed in HSC, MPP1, and MPP2 cells to be miR-221/222 targets, suggesting that a lot of the predicted and validated targets should not vulnerable to miR-221/222 motion in these cells or regulated on the degree of translation because it has been present with equipment [38]. Once more, even with Fos, not all unperturbed HSC or perturbed HSC present these modifications (Figs 1A and 2C). These restrictions within the motion of the miR-221/222 cluster stay to be investigated in larger element.

Three of the 6 IEGs selectively up-regulated in unperturbed hematopoiesis by miR-221/222 deficiency, specifically, Klf6, Nr4a1, and Zfp36 all would possibly cooperate to situation HSC for myeloid-granulocyte-biased hematopoiesis. Klf6, along with Runx1, promotes the transition of neutrophils from BM to blood [62,63].

Our discovering that miR-221/222 selectively up-regulates the expression of Klf6 in HSC, that miR-221/222-deficient mice have elevated ranges of granulocytes in spleen, and that serial transplantation of miR-221/222-deficient HSC results in the event of myeloid-granulocytic-biased HSC all might point out that this promotion to neutrophil growth begins in HSC in BM. Nr4a1, an orphan nuclear receptor, has been discovered expressed on myeloid-biased HSC [64]. Thus, miR-221/222 deficiency, selling the up-regulated expression of Nr4a1 (Fig 3A), would possibly cooperate with Klf6 to situation HSC for myeloid-granulocytic growth. Zfp36, an AU-rich RNA-binding protein [65,66], has been discovered to suppress hypoxia and cell cycle signaling, actions which might be recognized to affect HSC performances. In gene expression trajectories of neutrophil-granulocyte growth from HSC, Zfp36 has been recognized as a differentiation-determining issue [15,67,68]. It stays to be investigated how miR-221/222 deficiency focuses its impact in HSC to this selective set of genes, thereby conditioning them for myeloid-granulocytic growth [67].

A deeper understanding of the molecular mechanisms, how HSCs regulate engraftment in BM and stability self-renewal versus differentiation, has necessary medical implications for mobilizing protection towards infections, for bettering BM transplantations, and for preventing most cancers of hematopoietic precursor cells. Our findings recommend that expression of the miR-221/222 cluster safeguards long-lived, quiescent HSC from Fos/AP-1-induced activation ensuing within the discount of HSC, the proliferation of MPPs, and their differentiation and myeloid-granulocytic hematopoiesis [9,21,24,6971]. Protocols, which goal at activating miR-221/222 expression in HSC, could also be useful to keep up or reestablish hematopoietic efficiency by longevity, BM homing capability, quiescence, and myeloid-lymphoid multipotency over myeloid-biased hematopoiesis.

Supplies and strategies

Bone marrow and spleen preparation

BM and spleen-derived single-cell suspensions had been ready as described earlier [72]. With a view to reduce a possible continued “ex vivo” stimulation of gene expression, e.g., of IEG expression, which could happen throughout BM cell preparation, cell dealing with of the samples, which could affect the transcriptome analyses reported right here (qPCR and single-cell RNAseq), femurs and tibia from killed mice had been collected into tissue tradition medium containing 2 μg/ml Actinomycin-D. Actinomycin-D is an environment friendly RNA polymerase inhibitor and is used to reduce the uncontrolled “ex vivo” activation of gene expression modifications as additionally present in hematopoietic cells [73,74].

Move cytometry and cell sorting

We adhered to the rules for stream cytometry and cell sorting of hematopoietic stem and progenitor inhabitants and main lineage populations within the BM and the spleen as described earlier [72]. We used the nomenclature and gating technique of BM-derived stem and progenitor populations described by [57]. All gating technique, floor marker expression, and antibodies used within the research are listed in S5A–S5G Fig.

Quantitation of miR-221/222 expression on the single-cell degree

To measure the expression degree of miR-221 and miR-222 in single HSC, MPP1, and MPP2 populations, we mixed the protocols of [35,75,76] with slight modifications. Briefly, the one cells had been sorted in lysis buffer appropriate for the RT response after which 3-plex (miR-221, miR-222, and Sno202 particular) RT was carried out. Earlier than the standard simplex TaqMan qPCR, a 3-plexed cDNA preamplification PCR was performed. We included exterior calibration requirements consisting of 10-fold serial dilutions of artificial miRNA oligonucleotides of 105 to 100 copies (S6A and S6B Fig). Evaluation of those requirements revealed that assay effectivity diversified significantly and that sensitivity ranged between 100 and 102 molecules per response based mostly on no template management (NTC) (S6C and S6D Fig). Pre-BI cells of Pax5−/− mice expressing excessive ranges of miR-221 and miR-222 had been used as optimistic management whereas establishing the protocol. Variations in assay effectivity had been discovered unbiased of multiplexing degree and had been noticed even for serial dilutions of miRNA requirements in single-plex reactions; due to this fact, the Sno202 degree of single-cell samples not reaching the NTC had been thought-about as sorting failure and excluded from the evaluation (<10% of all sorted cells).

Single-cell RNA-library preparation and sequencing

Single-cell BM suspensions of unperturbed miR-221/222-proficient and miR-221/222-deficient mice had been stained for preparative cell sorting and MPP, CLP, linc-kit+Sca1, and linc-kitSca1 (5,200 every) cells (S5 Fig) had been utilized to the 10x Genomics workflow for cell capturing and era of scRNA gene expression (GEX) library utilizing the Chromium Single Cell 3′ Library & Gel Bead Package.

As a result of low cell counts of stem and progenitor cells, the BM-derived lineage-positive (B220+, CD3+, CD4+, CD8+, CD11b+, CD11c+,CD19+, Gr1+, NK1.1+, TER119+) cells had been first depleted on magnetic column (LS from Miltenyi), and the remaining lin single-cell suspensions from 4–4 perturbed or unperturbed miR-221/222-proficient and miR-221/222-deficient mice had been labeled individually for preparative FACS (S5 Fig), adopted by staining with TotalSeq-C anti-mouse Hashtag-1 or-4 (miR-221/222-proficient, perturbed: #1 or unperturbed: #4) or -Hashtag-2 or-5 (miR-221/222-deficient perturbed: #2 or unperturbed: #5) antibodies (BioLegend, #1: 155,861, #2: 155,863, #4: 155,867, #5: 155,869). The unperturbed or perturbed miR-221/222-proficient samples had been pooled with poor samples for scRNAseq. Then, 11,584 HSC, 12,605 MPP1, and 18,721 MPP2 sorted from pooled unperturbed samples or 8,900 HSC, 12,400 MPP1, and 9,100 MPP2 sorted from pooled perturbed samples had been then utilized to 10x Genomics workflow for cell capturing and scRNA gene expression (GEX) library preparation utilizing the Chromium Single Cell 5′ Library & Gel Bead Package in addition to the Single Cell 5′ Characteristic Barcode Library Package (10x Genomics). After cDNA amplification, the CiteSeq libraries had been ready individually utilizing the Single Index Package N Set A, whereas last GEX libraries had been obtained after fragmentation, adapter ligation, and last Index PCR utilizing the Single Index Package T Set A.

The BM-derived lineage-positive cells had been first depleted on magnetic column, and the remaining single-cell suspensions from 4 2-time transplanted animals had been stained for preparative sorting of CD45.2+ cells. After sorting 7,045 HSC, 6,363 MPP1, and three,532 MPP2 from miR-221/222-proficent HSC transplanted mice, the cells had been individually utilized, whereas 1,780 HSC, 771 MPP1, and 422 MPP2 sorted from poor HSC transplanted mice had been pooled after sorting and utilized to the 10x Genomics workflow utilizing the Chromium Single Cell 5′ Library & Gel Bead Package and Single Index Package T Set A.

For all libraries ready, the fragment sizes had been decided utilizing the Fragment Analyzer with the HS NGS Fragment Package (1 to six,000 bp) (Agilent), and library concentrations had been decided with Qubit HS DNA assay equipment (Life Applied sciences).

3′ GEX libraries and 5′ GEX+CITE libraries had been sequenced on a NextSeq500 machine (Illumina) utilizing Excessive Output v2 Kits (150 cycles) or on a NextSeq2000 machine (Illumina) utilizing both P2 reagents (200 cycles) or P3 reagents (200 cycles or 100 cycles) with the really helpful sequencing situations for (read1: 26 nt; read2: 98 nt; index1: 8 nt; index2: n.a.).

Single-cell transcriptome profiling

Uncooked indicators had been demultiplexed and transformed to fastq information utilizing DRAGEN (Ilumina). Detection of intact cells and expression quantification was carried out by cellranger (model 5.0.0) utilizing depend in default parameter settings with variety of anticipated cells set to three,000 and refdata-cellranger-mm10 as reference. Additional evaluation was performed in R (model 4.1.2) utilizing the Seurat bundle (model 4.0.5).

The mixing of the datasets from HSC, MPP1, MPP2, MPP [34], CLP, linc-kit+Sca1, and linc-kitSca1 cells of unperturbed miR-221/222-proficient and miR-221/222-deficient mice was carried out by following the mixing pipeline as described within the FindIntegrationAnchors (Seurat) R Documentation. Firstly, every library was log-normalized utilizing NormalizeData, 2,000 variable genes had been detected utilizing vst as choice methodology with FindVariableFeatures, variable genes had been scaled utilizing ScaleData, and 50 precept elements had been computed utilizing RunPCA. Subsequent, frequent anchors had been recognized by FindIntegrationAnchors utilizing rpca as discount, 2,000 anchor options, and 1:30 dimensions. Lastly, libraries had been merged utilizing IntegrateData.

The built-in information had been additional analyzed by a UMAP sequentially utilizing ScaleData, RunPCA for 50 precept elements, and RunUMAP utilizing 1:30 precept element. Transcriptionally related clusters (T-clusters) had been recognized by shared nearest neighbor (SNN) modularity optimization with FindNeighbors utilizing 1:30 precept elements and FindClusters with the resolutions of 0.2. The UMAP was evaluated by projection of scores for chosen developmental levels, outlined because the sum of log-normalized expression values.

Within the aggregated dataset, we detected a complete of 15 distinct clusters of BM lin populations (known as total- (T-) clusters). Excluding clusters representing lower than 5% of all analyzed cells, we additional investigated T-cluster 0 to 11. T-clusters 0, 2, and 10 might be characterised by the expression of HSC-related genes, largely from sorted HSC. T-clusters 3 and 4 had accrued expression of cell cycle–lively genes—cluster 3 for G1/S-phase and cluster 4 for G2/M part, largely from sorted MPP1-4 cells. T-cluster 5 contained T and NK lymphoid directed, T-cluster 6 contained B cell directed cells, largely from sorted CLP. T-cluster 8 contained erythropoiesis-directed, T-cluster 9 megakaryocyte-platelet-directed genes, and T-cluster 1 had accrued genes of earlier phases of myelopoiesis, largely from sorted linc-kit+Sca or linc-kitSca cells. Lastly, T-cluster 7 expressed genes of extra mature levels of basophil and mast cell growth, whereas in T-cluster 11, genes expressed in additional mature levels of neutrophils and granulocytes had been predominant. These T-clusters 7 and 11 consisted largely sorted linc-kit+Sca or linc-kitSca cells.

The identical workflow was used to merge HSC, MPP1, and MPP2 datasets. Specifically, the HSC, MPP1, and MPP2 from unperturbed and perturbed miR-221/222-proficient and miR-221/222-deficient mice, HSC, MPP1, and MPP2 sorted after the second transplantation of miR-221/222-proficient HSC in addition to the pool of HSC, MPP1, and MPP2 sorted after the second transplantation of miR-221/222-deficient HSC had been built-in, a UMAP was computed, and transcriptional cluster had been outlined utilizing the decision of 0.6. Clusters (E-clusters) with low high quality had been establish by visible inspection of UMI counts, variety of expressed, and proportion of mitochondrial genes. The contaminating cluster with erythrocytes contained cells with excessive Hbb expression and better UMI counts and variety of detected genes. Cells from low-quality clusters in addition to the contaminating cluster with lower than 5% contribution to all cells had been faraway from additional evaluation. Libraries with hashtag-labeled antibody stainings of miR-221/222-proficient and miR-221/222-deficient cells had been demultiplex by contrasting arcsinh remodeled hashtag 1 or 4 and hashtag 2 or 5 in a scatterplot and handbook gating.

Differential gene expression evaluation and comparability of variety of expressed genes and UMIs per cell had been carried out based mostly on downsampled libraries. In analogy to DESeq2 library dimension normalization, proposed for bulk sequencing [77], pseudo bulk samples had been created for every library after removing of low-quality and contaminating clusters by summing up the learn counts for every gene. Subsequent, the geometric imply was calculated for every gene expressed in a minimum of 75% of the thought-about cells in every library and a minimum of 75% of miR-221/222-proficient and miR-221/222-deficient cells within the swimming pools. The library dimension elements had been outlined because the median ratio between the UMI counts of the library and the corresponding geometric imply normalized to the variety of thought-about cells. Downsampling was carried out by subsampling of UMIs with sampling charges outlined because the ratios of the minimal and the respective dimension issue. Numbers of detected genes and UMIs had been computed after resembling the gene counts. Gene expression represents log2p-transformed UMI counts. Differential expression evaluation was carried out based mostly on subsampled however not normalized values. Specifically, for every gene log2p remodeled, all 0 values eliminated and a Mann–Whitney check was carried out. Subsequent, genes had been ranked by the fraction of expressing proficient (for miR-221/222-sensitive genes) or unperturbed (for perturbation-sensitive genes) cells, and a linear regression was carried out on the log2p fold change between miR-221/222-deficient and miR-221/222-proficient or fold change between perturbed and unperturbed samples (imply log2Exppoor-mean log2Expproficient or imply log2Expperturbed-mean log2Expunperturbed). The linear regression was carried out utilizing Prism (Model 9.0). Genes had been outlined as differentially expressed, if present in a minimum of 30% of the cells and the log2 fold change exceeding the 99% prediction band (PB).

Supporting data

S5 Fig. Gating technique and floor marker expression of stem and progenitor populations within the BM and lineage cells within the spleen.

After making ready single cell suspensions of (A) unperturbed or (B) perturbed miR-221/222-proficient or of (C) unperturbed or (D) perturbed miR-221/222-deficient BM, stem and progenitor cells had been stained for stream cytometry evaluation. (E) Single-cell suspension of miR-221/222-proficient or miR-221/222-deficient spleen or blood was ready, and the foremost hematopoietic lineage cells had been stained for stream cytometry evaluation. (F) Floor marker expression of the measured hematopoietic stem, progenitor, and lineage cells. HSC had been gated on lin (B220CD3CD4CD8CD19CD11cCD11bGr1NK1.1TER119) c-kit+Sca1+Flk2CD34CD150+CD48 cells. MPP1 was gated on linc-kit+Sca1+Flk2CD34+CD150+CD48 cells. MPP2 had been gated on linc-kit+Sca1+Flk2CD34+CD150+CD48+ cells. MPP3 had been gated on linc-kit+Sca1+Flk2CD34+CD150CD48+ cells. MPP4 had been gated on linc-kit+Sca1+Flk2+CD34+CD150CD48+ cells. CLPs had been gated on linc-kitloSca1loFlk2+IL7R+ cells. CMPs had been gated on linc-kit+Sca1CD34+CD16/32 cells. MEPs had been gated on linc-kit+Sca1CD34CD16/32 cells. GMPs had been gated on linc-kit+Sca1CD34+CD16/32+ cells. CD4 T cells had been gated as CD3+CD4+; CD8 T cells had been gated as CD3+CD8+ cell. B cells had been gated as CD4CD8B220+CD19+ cell. Myeloid cells had been gated as CD4CD8B220CD19CD11b+Gr1 cells. Granulocytes (Gran.) had been gated on CD4CD8B220CD19CD11b+Gr1+ cells. (G) Antibodies used within the analyses. The numerical information might be discovered within the FlowRepository.org by FR-FCM-Z6PS accession quantity. BM, bone marrow; CLP, frequent lymphoid progenitor; CMP, frequent myeloid progenitor; GMP, granulocyte-myelocyte progenitor; HSC, hematopoietic stem cell; MEP, megakaryocyte-erythroid progenitor; MPP, multipotent progenitor.

https://doi.org/10.1371/journal.pbio.3002015.s008

(TIF)

S6 Fig. Technique for the detection of copy numbers of miR-221/222 molecules in single cells.

(A) Round 105−100 copies of serially diluted artificial miR-221 oligonucleotide was measured 3 occasions (Run1-3) in 3 technical replicates. The TaqMan qPCR measurements had been performed after or with out (with out pre-amp) 10-cycle PRC amplification of the reverse transcript. (B) About 105−100 copies of serially diluted artificial miR-222 oligonucleotides had been measured 2 occasions (Run1 and a couple of) in 3 technical replicates. The TaqMan qPCR measurements had been performed after 10-cycle PRC amplification of the reverse transcript. (C) A complete of 45 single-cell sorted miR-221/222-proficient linc-kit+Sca1+CD150+CD48 (pool of HSC and MPP1) cells had been immediately sorted in lysis buffer, appropriate for miR-221- or miR-222-specific reverse transcription response. After 10-cycle PRC amplification of the reverse transcript, the copy numbers of miR-221 (purple) and miR-222 (blue) had been decided in particular person cells utilizing commonplace curves developed in the identical response plate. Purple and blue dots point out technical replicates (D) The restrict of detection in miR-221 and -222 copy numbers had been assigned by the very best worth of 10 NTCs, the place no cell was sorted within the lysis buffer. Purple and blue dots point out technical replicates. Pax5−/− pre-BI cells had been used as optimistic management. The imply of the technical replicates are calculated and plotted for 10 particular person cells. The numerical information might be present in S1 Knowledge. HSC, hematopoietic stem cell; MPP, multipotent progenitor; NTC, no template management.

https://doi.org/10.1371/journal.pbio.3002015.s009

(TIF)

References

  1. 1.
    Seita J, Weissman IL. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip Rev Syst Biol Med. 2010;2(6):640–653. pmid:20890962
  2. 2.
    Morrison SJ, Scadden DT. The bone marrow area of interest for haematopoietic stem cells. Nature. 2014;505(7483):327–334. pmid:24429631
  3. 3.
    Busch Okay, Klapproth Okay, Barile M, Flossdorf M, Holland-Letz T, Schlenner SM, et al. Basic properties of unperturbed haematopoiesis from stem cells in vivo. Nature. 2015;518(7540):542–546. pmid:25686605
  4. 4.
    Hofer T, Busch Okay, Klapproth Okay, Rodewald HR. Destiny Mapping and Quantitation of Hematopoiesis In Vivo. Annu Rev Immunol. 2016;34:449–478. pmid:27168243
  5. 5.
    Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W, Jaworski M, et al. Hematopoietic stem cells reversibly change from dormancy to self-renewal throughout homeostasis and restore. Cell. 2008;135(6):1118–1129. pmid:19062086
  6. 6.
    Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. SLAM household receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121(7):1109–1121. pmid:15989959
  7. 7.
    Cabezas-Wallscheid N, Klimmeck D, Hansson J, Lipka DB, Reyes A, Wang Q, et al. Identification of regulatory networks in HSCs and their fast progeny through built-in proteome, transcriptome, and DNA methylome evaluation. Cell Stem Cell. 2014;15(4):507–522. pmid:25158935
  8. 8.
    Pei W, Shang F, Wang X, Fanti AK, Greco A, Busch Okay, et al. Resolving Fates and Single-Cell Transcriptomes of Hematopoietic Stem Cell Clones by PolyloxExpress Barcoding. Cell Stem Cell. 2020;27(3):383–395 e8. pmid:32783885
  9. 9.
    Pei W, Feyerabend TB, Rossler J, Wang X, Postrach D, Busch Okay, et al. Polylox barcoding reveals haematopoietic stem cell fates realized in vivo. Nature. 2017;548(7668):456–460. pmid:28813413
  10. 10.
    Halliwell B. Cell tradition, oxidative stress, and antioxidants: avoiding pitfalls. Biomed J. 2014;37(3):99–105. pmid:24923566
  11. 11.
    Lan Q, Mercurius KO, Davies PF. Stimulation of transcription elements NF kappa B and AP1 in endothelial cells subjected to shear stress. Biochem Biophys Res Commun. 1994;201(2):950–956. pmid:8003036
  12. 12.
    McKim DB, Yin W, Wang Y, Cole SW, Godbout JP, Sheridan JF. Social Stress Mobilizes Hematopoietic Stem Cells to Set up Persistent Splenic Myelopoiesis. Cell Rep. 2018;25(9):2552–2562 e3. pmid:30485819
  13. 13.
    Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to power an infection. Nature. 2010;465(7299):793–797. pmid:20535209
  14. 14.
    Bowman TV. Attending to the Ncor of HSC emergence. Blood. 2014;124(10):1541–1542. pmid:25190746
  15. 15.
    Khan N, Downey J, Sanz J, Kaufmann E, Blankenhaus B, Pacis A, et al. M. tuberculosis Reprograms Hematopoietic Stem Cells to Restrict Myelopoiesis and Impair Skilled Immunity. Cell. 2020;183(3):752–770 e22.
  16. 16.
    Larsson J, Scadden D. Nervous exercise in a stem cell area of interest. Cell. 2006;124(2):253–255. pmid:16439198
  17. 17.
    Clapes T, Lefkopoulos S, Trompouki E. Stress and Non-Stress Roles of Inflammatory Alerts throughout HSC Emergence and Upkeep. Entrance Immunol. 2016;7:487. pmid:27872627
  18. 18.
    Passegue E, Ernst P. IFN-alpha wakes up sleeping hematopoietic stem cells. Nat Med. 2009;15(6):612–613. pmid:19498372
  19. 19.
    Matatall KA, Shen CC, Challen GA, King KY. Sort II interferon promotes differentiation of myeloid-biased hematopoietic stem cells. Stem Cells. 2014;32(11):3023–3030. pmid:25078851
  20. 20.
    Panopoulos AD, Watowich SS. Granulocyte colony-stimulating issue: molecular mechanisms of motion throughout regular state and ’emergency’ hematopoiesis. Cytokine. 2008;42(3):277–288. pmid:18400509
  21. 21.
    Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol. 2014;14(5):302–314. pmid:24751955
  22. 22.
    Geiger H, de Haan G, Florian MC. The ageing haematopoietic stem cell compartment. Nat Rev Immunol. 2013;13(5):376–389. pmid:23584423
  23. 23.
    Pang WW, Value EA, Sahoo D, Beerman I, Maloney WJ, Rossi DJ, et al. Human bone marrow hematopoietic stem cells are elevated in frequency and myeloid-biased with age. Proc Natl Acad Sci U S A. 2011;108(50):20012–20017. pmid:22123971
  24. 24.
    Dorshkind Okay, Hofer T, Montecino-Rodriguez E, Pioli PD, Rodewald HR. Do haematopoietic stem cells age? Nat Rev Immunol. 2020;20(3):196–202. pmid:31740804
  25. 25.
    Bartel DP. MicroRNAs: goal recognition and regulatory capabilities. Cell. 2009;136(2):215–233. pmid:19167326
  26. 26.
    Fabian MR, Sonenberg N, Filipowicz W. Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 2010;79:351–379. pmid:20533884
  27. 27.
    Rana TM. Illuminating the silence: understanding the construction and performance of small RNAs. Nat Rev Mol Cell Biol. 2007;8(1):23–36. pmid:17183358
  28. 28.
    Montagner S, Deho L, Monticelli S. MicroRNAs in hematopoietic growth. BMC Immunol. 2014;15:14. pmid:24678908
  29. 29.
    Bissels U, Bosio A, Wagner W. MicroRNAs are shaping the hematopoietic panorama. Haematologica. 2012;97(2):160–167. pmid:22058204
  30. 30.
    Wilczynska A, Bushell M. The complexity of miRNA-mediated repression. Cell Dying Differ. 2015;22(1):22–33. pmid:25190144
  31. 31.
    Guo S, Lu J, Schlanger R, Zhang H, Wang JY, Fox MC, et al. MicroRNA miR-125a controls hematopoietic stem cell quantity. Proc Natl Acad Sci U S A. 2010;107(32):14229–14234. pmid:20616003
  32. 32.
    O’Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Entrance Endocrinol (Lausanne). 2018;9:402. pmid:30123182
  33. 33.
    Luinenburg DG, de Haan G. MicroRNAs in hematopoietic stem cell growing old. Mech Ageing Dev. 2020;189:111281. pmid:32512019
  34. 34.
    Wang P, Liu XM, Ding L, Zhang XJ, Ma ZL. mTOR signaling-related MicroRNAs and Most cancers involvement. J Most cancers. 2018;9(4):667–673. pmid:29556324
  35. 35.
    Petriv OI, Kuchenbauer F, Delaney AD, Lecault V, White A, Kent D, et al. Complete microRNA expression profiling of the hematopoietic hierarchy. Proc Natl Acad Sci U S A. 2010;107(35):15443–15448. pmid:20702766
  36. 36.
    Knoll M, Simmons S, Bouquet C, Grun JR, Melchers F. miR-221 redirects precursor B cells to the BM and regulates their residence. Eur J Immunol. 2013;43(9):2497–2506. pmid:23716169
  37. 37.
    Crisafulli L, Muggeo S, Uva P, Wang Y, Iwasaki M, Locatelli S, et al. MicroRNA-127-3p controls murine hematopoietic stem cell upkeep by limiting differentiation. Haematologica. 2019;104(9):1744–1755. pmid:30792210
  38. 38.
    Felli N, Fontana L, Pelosi E, Botta R, Bonci D, Facchiano F, et al. MicroRNAs 221 and 222 inhibit regular erythropoiesis and erythroleukemic cell progress through equipment receptor down-modulation. Proc Natl Acad Sci U S A. 2005;102(50):18081–18086. pmid:16330772
  39. 39.
    Petkau G, Kawano Y, Wolf I, Knoll M, Melchers F. MiR221 promotes precursor B-cell retention within the bone marrow by amplifying the PI3K-signaling pathway in mice. Eur J Immunol. 2018;48(6):975–989. pmid:29505092
  40. 40.
    de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, et al. Transgenic mice with hematopoietic and lymphoid particular expression of Cre. Eur J Immunol. 2003;33(2):314–325. pmid:12548562
  41. 41.
    Mikami Y, Philips RL, Sciume G, Petermann F, Meylan F, Nagashima H, et al. MicroRNA-221 and -222 modulate intestinal inflammatory Th17 cell response as detrimental suggestions regulators downstream of interleukin-23. Immunity. 2021;54(3):514–525 e6. pmid:33657395
  42. 42.
    Wigton EJ, Mikami Y, McMonigle RJ, Castellanos CA, Wade-Vallance AK, Zhou SK, et al. MicroRNA-directed pathway discovery elucidates an miR-221/222-mediated regulatory circuit in school change recombination. J Exp Med. 2021;218(11). pmid:34586363
  43. 43.
    Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, et al. DIANA-TarBase v8: a decade-long assortment of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018;46(D1):D239–D245. pmid:29156006
  44. 44.
    McGeary SE, Lin KS, Shi CY, Pham TM, Bisaria N, Kelley GM, et al. The biochemical foundation of microRNA focusing on efficacy. Science. 2019;366(6472). pmid:31806698
  45. 45.
    Chen Y, Wang X. miRDB: a web-based database for prediction of purposeful microRNA targets. Nucleic Acids Res. 2020;48(D1):D127–D131. pmid:31504780
  46. 46.
    Errico MC, Felicetti F, Bottero L, Mattia G, Boe A, Felli N, et al. The abrogation of the HOXB7/PBX2 complicated induces apoptosis in melanoma by the miR-221&222-c-FOS pathway. Int J Most cancers. 2013;133(4):879–892.
  47. 47.
    Desterke C, Bennaceur-Griscelli A, Turhan AG. EGR1 dysregulation defines an inflammatory and leukemic program in cell trajectory of human-aged hematopoietic stem cells (HSC). Stem Cell Res Ther. 2021;12(1):419. pmid:34294125
  48. 48.
    Angel P, Karin M. The position of Jun, Fos and the AP-1 complicated in cell-proliferation and transformation. Biochim Biophys Acta. 1991;1072(2–3):129–157. pmid:1751545
  49. 49.
    Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment evaluation: a knowledge-based method for decoding genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–15550. pmid:16199517
  50. 50.
    Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, et al. PGC-1alpha-responsive genes concerned in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34(3):267–273. pmid:12808457
  51. 51.
    Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, et al. Dimensionality discount for visualizing single-cell information utilizing UMAP. Nat Biotechnol. 2019;37:38–44.
  52. 52.
    Dahlin JS, Hamey FK, Pijuan-Sala B, Shepherd M, Lau WWY, Nestorowa S, et al. A single-cell hematopoietic panorama resolves 8 lineage trajectories and defects in Package mutant mice. Blood. 2018;131(21):e1–e11. pmid:29588278
  53. 53.
    Wey S, Luo B, Lee AS. Acute inducible ablation of GRP78 reveals its position in hematopoietic stem cell survival, lymphogenesis and regulation of stress signaling. PLoS ONE. 2012;7(6):e39047. pmid:22723926
  54. 54.
    Schappi JM, Krbanjevic A, Rasenick MM. Tubulin, actin and heterotrimeric G proteins: coordination of signaling and construction. Biochim Biophys Acta. 2014;1838(2):674–681. pmid:24071592
  55. 55.
    Wierenga AT, Vellenga E, Schuringa JJ. Convergence of hypoxia and TGFbeta pathways on cell cycle regulation in human hematopoietic stem/progenitor cells. PLoS ONE. 2014;9(3):e93494.
  56. 56.
    Chan KYY, Zhang C, Wong YTS, Zhang XB, Wang CC, Ng WH, et al. R4 RGS proteins suppress engraftment of human hematopoietic stem/progenitor cells by modulating SDF-1/CXCR4 signaling. Blood Adv. 2021;5(21):4380–4392. pmid:34500454
  57. 57.
    Murdaugh RL, Hoegenauer KA, Kitano A, Holt MV, Hill MC, Shi X, et al. The histone H3.3 chaperone HIRA restrains erythroid-biased differentiation of grownup hematopoietic stem cells. Stem Cell Reviews. 2021;16(8):2014–2028. pmid:34242617
  58. 58.
    Zhang X, Kluger Y, Nakayama Y, Poddar R, Whitney C, DeTora A, et al. Gene expression in mature neutrophils: early responses to inflammatory stimuli. J Leukoc Biol. 2004;75(2):358–372. pmid:14634056
  59. 59.
    Paul F, Arkin Y, Giladi A, Jaitin DA, Kenigsberg E, Keren-Shaul H, et al. Transcriptional Heterogeneity and Lineage Dedication in Myeloid Progenitors. Cell. 2015;163(7):1663–1677. pmid:26627738
  60. 60.
    Ericson JA, Duffau P, Yasuda Okay, Ortiz-Lopez A, Rothamel Okay, Rifkin IR, et al. Gene expression through the era and activation of mouse neutrophils: implication of novel purposeful and regulatory pathways. PLoS ONE. 2014;9(10):e108553. pmid:25279834
  61. 61.
    Kim MH, Yang D, Kim M, Kim SY, Kim D, Kang SJ. A late-lineage murine neutrophil precursor inhabitants displays dynamic modifications throughout demand-adapted granulopoiesis. Sci Rep. 2017;7:39804. pmid:28059162
  62. 62.
    Khoyratty TE, Ai Z, Ballesteros I, Eames HL, Mathie S, Martin-Salamanca S, et al. Distinct transcription issue networks management neutrophil-driven irritation. Nat Immunol. 2021;22(9):1093–1106. pmid:34282331
  63. 63.
    Chevre R, Soehnlein O. Neutrophil life in three acts: a manufacturing by completely different stage administrators. Nat Immunol. 2021;22(9):1072–1074. pmid:34326535
  64. 64.
    Land RH, Rayne AK, Vanderbeck AN, Barlowe TS, Manjunath S, Gross M, et al. The orphan nuclear receptor NR4A1 specifies a definite subpopulation of quiescent myeloid-biased long-term HSCs. Stem Cells. 2015;33(1):278–288. pmid:25284014
  65. 65.
    Baou M, Norton JD, Murphy JJ. AU-rich RNA binding proteins in hematopoiesis and leukemogenesis. Blood. 2011;118(22):5732–5740. pmid:21917750
  66. 66.
    Loh XY, Solar QY, Ding LW, Mayakonda A, Venkatachalam N, Yeo MS, et al. RNA-Binding Protein ZFP36L1 Suppresses Hypoxia and Cell-Cycle Signaling. Most cancers Res. 2020;80(2):219–233. pmid:31551365
  67. 67.
    Quick EM, Sporrij A, Manning M, Rocha EL, Yang S, Zhou Y, et al. Exterior indicators regulate steady transcriptional states in hematopoietic stem cells. Elife. 2021;10. pmid:34939923
  68. 68.
    Lummertz da Rocha E, Rowe RG, Lundin V, Malleshaiah M, Jha DK, Rambo CR, et al. Reconstruction of complicated single-cell trajectories utilizing CellRouter. Nat Commun. 2018;9(1):892. pmid:29497036
  69. 69.
    Tullai JW, Schaffer ME, Mullenbrock S, Sholder G, Kasif S, Cooper GM. Fast-early and delayed major response genes are distinct in operate and genomic structure. J Biol Chem. 2007;282(33):23981–23995. pmid:17575275
  70. 70.
    Nakamura-Ishizu A, Takizawa H, Suda T. The evaluation, roles and regulation of quiescence in hematopoietic stem cells. Growth. 2014;141(24):4656–4666. pmid:25468935
  71. 71.
    Borghesi L. Hematopoiesis in steady-state versus stress: self-renewal, lineage destiny selection, and the conversion of hazard indicators into cytokine indicators in hematopoietic stem cells. J Immunol. 2014;193(5):2053–2058. pmid:25128551
  72. 72.
    Cossarizza A, Chang HD, Radbruch A, Abrignani S, Addo R, Akdis M, et al. Pointers for the usage of stream cytometry and cell sorting in immunological research (third version). Eur J Immunol. 2021;51(12):2708–3145. pmid:34910301
  73. 73.
    Westendorf Okay, Okhrimenko A, Grun JR, Schliemann H, Chang HD, Dong J, et al. Unbiased transcriptomes of resting human CD4(+) CD45RO(+) T lymphocytes. Eur J Immunol. 2014;44(6):1866–1869.
  74. 74.
    Zuckerman KS, Sullivan R, Quesenberry PJ. Results of actinomycin D in vivo on murine erythroid stem cells. Blood. 1978;51(5):957–969. pmid:638254
  75. 75.
    Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, et al. Actual-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 2005;33(20):e179. pmid:16314309
  76. 76.
    Tang F, Hajkova P, Barton SC, O’Carroll D, Lee C, Lao Okay, et al. 220-plex microRNA expression profile of a single cell. Nat Protoc. 2006;1(3):1154–1159. pmid:17406397
  77. 77.
    Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq information with DESeq2. Genome Biol. 2014;15(12):550. pmid:25516281
  78. 78.
    Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression information. Nat Biotechnol. 2015;33(5):495–502. pmid:25867923
  79. 79.
    Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar Okay, Goldman M, et al. Extremely Parallel Genome-wide Expression Profiling of Particular person Cells Utilizing Nanoliter Droplets. Cell. 2015;161(5):1202–1214. pmid:26000488
  80. 80.
    Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, third, et al. Complete Integration of Single-Cell Knowledge. Cell. 2019;177(7):1888–1902 e21. pmid:31178118
  81. 81.
    Hao Y, Hao S, Andersen-Nissen E, Mauck WM, third, Zheng S, Butler A, et al. Built-in evaluation of multimodal single-cell information. Cell. 2021;184(13):3573–3587 e29. pmid:34062119

[ad_2]

LEAVE A REPLY

Please enter your comment!
Please enter your name here