Introduction
Tissue stem cells have a self-renewal capacity and multipotency to maintain adult tissue homeostasis and repair stress-induced tissue damage (Wagers and Weissman,
2004; Goodell and Rando,
2015). Stress is the disruption of homeostasis through changes in signaling, epigenomic, and transcriptional mechanisms mediated by extrinsic stimuli and intrinsic factors, resulting in transient or irreversible tissue damage, such as aging-associated diseases including cancer (Oh et al,
2014; López-Otín et al,
2023). Hematopoietic stem cells (HSCs) respond to various stresses, such as infection, inflammation, and myeloablation, and expand hematopoietic stem and progenitor cells (HSPCs) to produce mature blood cells (Essers et al,
2009; Baldridge et al,
2010; Haas et al,
2015); however, the mechanisms by which HSCs maintain hematopoiesis based on their self-renewal and differentiation fates in differential responses to homeostatic and stress conditions have yet to be elucidated.
High mobility group AT-hook 2 (Hmga2), a chromatin modifier protein, is highly expressed in fetal HSCs and is necessary for an enhanced self-renewal capacity (Copley et al,
2013). Hmga2 activates the transcription of target genes by competing with the linker histone H1 and/or binding to the regulatory regions of genes via its AT-hook domains (Fusco and Fedele,
2007). The overexpression of
Hmga2 via a retrovirus vector or a transgene was shown to enhance the self-renewal capacity of adult HSCs and the megakaryocyte and erythroid differentiation in humans and mice under in vivo conditions (Copley et al,
2013; Rowe et al,
2016; Kumar et al,
2019), but was insufficient to induce leukemic transformation (Ikeda et al,
2011; Bai et al,
2021). We generated
Rosa26 locus
Hmga2 conditional knock-in (KI) mice and recently reported that the overexpression of
Hmga2 enhanced the self-renewal capacity of adult HSCs and maintained their fitness in the bone marrow (BM) of both primary mice and transplanted mice (Sun et al,
2022), while lethal irradiation followed by BM transplantation induced the excessive replication of HSCs and systemic inflammation and subsequently depleted wild-type (WT) HSCs in the long term (Rodrigues-Moreira et al,
2017; Caiado et al,
2021).
In the present study, we generated new
Hmga2 conditional knock-out (cKO) mice and examined normal and stress hematopoiesis using both
Hmga2 cKO and KI mice. Under homeostatic conditions, neither
Hmga2 cKO nor KI mice showed a significant change in hematopoietic phenotypes; however, in response to an injection of 5-fluorouracil (5-FU), the proliferation of HSCs was enhanced in
Hmga2 KI mice and hematopoietic recovery was rapid, whereas HSC and megakaryocyte progenitor (MkP) cell numbers significantly decreased in
Hmga2 cKO mice and hematopoietic recovery was delayed. Since the Hmga2 gene appeared to repress the transcription of interferon-stimulated genes and inflammatory response genes in HSCs, we assessed Hmga2-binding regions and chromatin accessibility in cells after the in vivo injection of 5-FU or an in vitro treatment with TNF-α, a critical cytokine in inflammatory responses (Pronk et al,
2011; Yamashita and Passegué,
2019). We found that Hmga2 bound to distinct regions and reduced chromatin accessibility in inflammation regulator genes, such as the Rfx5 transcription factor (TF), after these stress insults. Since the acidic domain in the Hmga2 protein was directly phosphorylated by Casein kinase 2 (CK2) (Sgarra et al,
2009), which has been implicated in the modulation of the TNF-α-NF-kB pathway (Borgo et al,
2021), the inhibition of phosphorylation in the acidic domain by a substitution with alanine impaired the stress-induced chromatin binding of Hmga2, chromatin accessibility, the transcription of target genes, and the expansion of HSCs under stress conditions. While the
Hmga2 gene did not appear to be necessary for adult hematopoiesis under homeostatic conditions, we herein elucidated the molecular mechanisms by which HSCs drove hematopoietic regeneration under stress conditions by activating the expression and function of Hmga2.
Discussion
We demonstrated that Hmga2 was vitalized by the TNF-α-CK2 signaling for the expansion of HSCs to conduct stress hematopoiesis. Hmga2 are involved in regulation of chromatin accessibility and the transcriptome downstream of the TNF-α-CK2 axis in HSCs. Under stress conditions, such as transplantation and after the 5-FU injection, which elevated inflammatory cytokines in BM plasma cells
Hmga2 KI mice showed the enhanced expansion of HSCs and rapid hematopoietic recovery increasing megakaryocyte-biased HSCs and MkP cells in BM and platelets in PB. In contrast, after the 5-FU injection,
Hmga2 cKO mice decreased the number of HSCs and showed delayed hematopoietic recovery, which resulted in the death of
Hmga2 cKO mice, suggesting that the presence of
Hmga2 was critical for stress hematopoiesis. In fact, adult HSCs increased the expression of the endogenous Hmga2 at the protein, but not mRNA level in response to stress stimuli, including TNF-α, which induced the phosphorylation of the Hmga2 protein at its acidic domain due to the activation of CK2 kinase, thereby forming the TNF-α-CK2-Hmga2 axis. In contrast, under homeostatic conditions, neither
Hmga2 cKO nor
Hmga2 KI mice showed a significant change in hematopoietic phenotypes, indicating that the
Hmga2 gene was not necessary for normal steady-state hematopoiesis. The present results revealed a role for the TNF-α-CK2-Hmga2 axis in the expansion and survival of HSCs and the regeneration of hematopoiesis driving megakaryopoiesis under stress conditions (a model is shown in Fig.
7B).
A stimulation with TNF-α was considered to be detrimental for HSCs (Pronk et al,
2011; Ishida et al,
2017), whereas the TNF-α-mediated activation of NF-kB was recently shown to prevent HSC necroptosis (Yamashita and Passegué,
2019). In development, sterile TNF-α signaling is required for the generation of HSCs (Espín-Palazón et al,
2014). Therefore, the mechanisms by which a stimulation with TNF-α regulates the transcription of downstream genes and the behavior of HSCs in a context-dependent manner remain unclear. The present study revealed that under stress conditions, including a TNF-α stimulation, the expression of
Hmga2 increased the expansion of HSCs, but not MPP cells, and drove megakaryopoiesis. In this process, the Hmga2 protein was phosphorylated by CK2 downstream of TNF-α, thereby regulating the chromatin accessibility and chromatin binding of TFs, demonstrating critical roles for the TNF-α-CK2-Hmga2 axis in stress hematopoiesis. NF-kB TF has been implicated in the activation of
Igf2bp2/Imp2 transcription in mouse embryonic fibroblast cells in cooperation with Hmga2 (Cleynen et al,
2007), suggesting a link between these genes and transcriptional regulation in HSCs mediated by TNF-α. The overexpression of
Hmga2 in WT adult HSCs is not sufficient for the development of malignancy in mice and non-human primates (Kumar et al,
2019; Bonner et al,
2021; Sun et al,
2022) and also presumably in humans after gene therapies (Cavazzana-Calvo et al,
2010). Since TNF-α levels are constitutively elevated in aged humans and patients with MDS (Sawanobori et al,
2003; Vasto et al,
2007), the TNF-α-CK2-Hmga2 axis may increase the expression level of
HMGA2 and contribute to the progression of MDS HSCs that harbor driver mutations. Further studies are needed to clarify whether Hmga2 and NF-kB are integrated to regulate transcription for the expansion and survival of normal and malignant HSCs under stress conditions.
CK2 is a critical kinase that regulates various cellular functions in embryos and adult tissues, and its enhanced activation may impair the functions of tissues and lead to cancer development (Borgo et al,
2021). While the AT-hooks of Hmga2 are required for DNA-binding and transcription-activating properties, the phosphorylation of Hmga2 at the C terminus was shown to be mediated by CK2 and affected its DNA-binding capacity in in vitro settings (Sgarra et al,
2009; Maurizio et al,
2011). We consistently confirmed that the acidic domain in the Hmga2 protein was directly phosphorylated by CK2. Furthermore, the phosphorylation of the Hmga2 protein was involved in TNF-α-induced chromatin accessibility to close the binding sites of the Rfx5 TF, which is expressed in HSCs and immune cells, and was also shown to regulate the expression of MHC class II genes (Clausen et al,
1998). The transduction of
Rfx5 decreased the numbers of WT and
Hmga2 KI HSCs after the stimulation with TNF-α, suggesting that the suppression of Rfx5 function was critical for the survival of HSCs under stress conditions. In addition to the phosphorylation-dependent property of inhibiting the binding of Rfx5, under stress conditions, Hmga2 opened the binding sites of GATA family genes and closed those of AP-1 family genes in HSCs in a manner that dwas not dependent on the phosphorylation of Hmga2, and also regulated the transcription of their target genes, which may contribute to the expansion of HSCs and promotion of megakaryopoiesis (Fig.
7B). Although the Hmga2 protein has been proposed to facilitate the chromatin binding of other TFs through their direct association (Fusco and Fedele,
2007), Hmga2-ChIP-seq revealed enriched AT-rich sequences via the AT-hook of Hmga2, but no enriched TF-binding motifs in the Hmga2-binding regions in blood cells, which were mostly non-regulatory regions of genes lacking active histone modifications, such as H3K27ac and H3K4me1. Further studies are needed to elucidate the molecular mechanisms by which the C-terminal phosphorylation of Hmga2 modulates its chromatin binding to non-regulatory regions and simultaneously regulates the transcription of target genes mediated by TFs. Nevertheless, the present results suggest critical roles for the acidic domain and its phosphorylation in Hmga2 in the regulation of chromatin accessibility to modulate the transcription for HSCs in stress conditions.
The present study has an important limitation. Our mouse experiments were mostly performed under transplantation and tamoxifen injection settings, which potentially cause severe stress for HSCs, such as increased inflammatory environments in BM and the homing and replication of HSCs. We noted that after the 5-FU injection, the recoveries of progenitor cells and platelets, but not HSCs, were slower in transplanted mice following the tamoxifen injection than in primary mice. Although we compared data from our mouse experiments between WT and Hmga2 mutant mice subjected to the same transplantation conditions, these manipulations may mask and/or exacerbate the property of Hmga2 and the functionality of HSCs, which may affect our interpretation of the results obtained. Further studies are needed to clarify the role of Hmga2 in HSCs under natural conditions without transplantation.
We demonstrated that Hmga2 dynamically regulated chromatin and the transcription of genes in HSCs accompanied with the phosphorylation of the Hmga2 protein, which affects the fate of HSCs under stress conditions. The present results provide a possibility for therapeutic interventions based on the TNF-α-CK2-Hmga2 axis to promote the regeneration of normal hematopoiesis, but also prevent the expansion of MDS stem cells in patients.
Methods
Mice
All mice were of the C57BL/6 background.
Rosa26-flox-stop-flox-HA-Hmga2-eGFP conditional KI mice were generated as previously reported (Sun et al,
2022), and were crossed with
Rosa26-Cre-ERT2 KI mice (TaconicArtemis).
Hmga2flox/flox cKO mice were generated using murine C57BL/6 ES cells and the
Hmga2 Exon 2 and Exon 3-fused mini-gene with loxP sequences ligated into the pFFRT-PGKneo vector.
Hmga2flox/flox KO mice were crossed with
Cre-ERT2 mice and
Vav-iCre mice. C57BL/6 mice congenic for the
Ly5 locus (CD45.1) were purchased from Sankyo-Lab Service. Adult male or female mice (8–12 weeks) were used as donors and recipients for experiments. Two milligrams of tamoxifen (T5648, Sigma-Aldrich) was administered via an intraperitoneal injection for 5 consecutive days to overexpress and deletion of
Hmga2. All experiments using these mice were performed in accordance with our institutional guidelines for the use of laboratory animals and approved by the Review Board for Animal Experiments of Kumamoto University (Kumamoto, Japan). Reference numbers were A2020-050 and A2022-022. All mouse experiments were performed without randomization and blinding.
Cells
293T and 293GPG cell lines were cultured in DMEM containing 10% fetal bovine serum (Ory et al,
1996). Mycoplasma contamination was tested in all cell lines by performing PCR.
Flow cytometry and antibodies
Flow cytometry and cell sorting were performed using the following anti-murine antibodies purchased from BioLegend or eBioscience (clone and catalog numbers): CD45.2 (104, 109820), CD45.1 (A20, 110730), Gr-1 (RB6-8C5, 108404), Mac1 (M1/70, 101208), Ter119 (116204), IL-7Rα (A7R34, 121104), B220 (RA3-6B2, 103212), CD3e (145-2C11, 100304), CD4 (L3T4, 100526), CD8α (53-6.7, 100714), c-Kit (2B8, 105812), Sca-1 (D7, 108114), FcγRII-III (93, 101308), CD34 (MEC14.7, 11-0341-85), CD41 (MWReg3, 133928), CD48 (HM48-1, 103443), CD105 (MJ7/18, 120414), CD135 (A2F10, 135306), CD150 (TC15-12F12.2, 115924), and CD201 (eBio1560, 2071517). The lineage mixture solution contained biotin-conjugated anti-Gr-1, B220, CD3e, CD8α, Ter119, and IL-7Rα antibodies. All analyses and cell sorting were performed as described in our previous study (Yokomizo-Nakano et al,
2023).
Virus vectors and virus transduction
Mouse
Rfx5, Rfx3,
Hmga2, and
Hmga2-5A lacking 3′UTR were subcloned into the pGCDN-sam-ires-NGFR vector. A VSV-G pseudo-type retroviral supernatant was prepared by transfecting a vector plasmid into 293T cells or 293GPG packaging cells. Virus transduction was performed as described in our previous study (Yokomizo-Nakano et al,
2020). Briefly, purified HSPCs were incubated in serum-free SF-03 medium supplemented with 0.1% BSA, 50 μM 2-ME, 10 ng/mL mouse SCF, and 100 ng/mL mouse TPO for 24 h and then transduced with the indicated virus vectors in the presence of 1 μg/mL RetroNectin and 10 μg/mL protamine sulfate.
Liquid culture assay
One day after virus transduction, the liquid culture assay was performed after the infection medium was replaced with SF-03 medium supplemented with 0.1% BSA, 50 μM 2-ME, 10 ng/ml mouse SCF, and 100 ng/ml murine TPO. Cells were treated with 100 ng/ml TNF-α (SBI). All in vitro experiments were conducted without randomization or blinding.
In vitro kinase assay
An in vitro kinase assay was performed as previously reported (Kubota et al,
2013). Cell lysates expressing FLAG-Hmga2 or FLAG-Hmga2-5A were immunoprecipitated by anti-FLAG antibody-precoated protein G beads and then incubated with CK2 (NEB) in protein kinase buffer containing 100 M unlabeled ATP at 30 °C for 30 min. Phosphorylated bands were immunoblotted using Phos-tag (Fujifilm).
CytoTell labeling assay
The CytoTell labeling assay was performed as previously reported (Umemoto et al,
2022). Briefly, purified stem cells were stained with CytoTellTM Red 650 (AAT Bioquest) diluted at 1/500 in PBS for 10 min. Stained cells were cultured or transplanted into 5-FU-treated recipient mice and then analyzed 2.5 days after transplantation.
Cytokine array
Cytokine levels in BM were measured by the Proteome Profiler Mouse Cytokine Array Kit, Panel A (ARY006, R and D Systems). BM was flushed out by 500 μl of PBS and centrifuged at 2000 × g for 15 min. Supernatants were applied to a cytokine array.
Assay for transposase-accessible chromatin using sequencing (ATAC-seq)
ATAC-seq libraries were prepared using the Omni-ATAC protocol (Corces et al,
2017). Briefly, cells were lysed and then incubated with transposase solution containing 1 μL of Tagment DNA Enzyme (Illumina). Transposed DNA was purified and then amplified using a NEBNext High Fidelity 2× PCR Master Mix (New England Biolabs) with indexed primers. The prepared libraries were sequenced on NextSeq500 (Illumina) with 38-bp paired-end reads. Bowtie2 (v2.2.6) was used to map reads to the reference genome (UCSC/mm9). Peak calling and motif analyses were performed using HOMER (v4.9). Heatmap clustering was conducted by deepTools (v3.5.0). Transcription factor footprints were analyzed using Transcription factor Occupancy prediction By Investigation of ATAC-seq Signal (TOBIAS, version 0.15.1) (Bentsen et al,
2020).
Chromatin immunoprecipitation sequencing (ChIP-seq)
ChIP-seq was performed as described in our previous study (Yokomizo-Nakano et al,
2020). Briefly, cells were fixed by 1.0% paraformaldehyde at 37 °C for 5 min. Fixed cells were lysed and sonicated 15 times at an amplitude of 50% for 10 sec. Samples were incubated with an anti-FLAG antibody (Sigma M2) or anti-HA antibody (Abcam, ab9110) conjugated by Dynabeads protein A/G at 4 °C overnight. ChIP-seq libraries were generated using the ThruPLEX DNA-seq kit (Rubicon Genomics) and then sequenced on NextSeq500 (Illumina). Bowtie2 (v2.2.6) was used to map reads to the reference genome (UCSC/mm9). Peak calling and motif analyses were performed using HOMER (v4.9). Heatmap clustering was conducted by deepTools (v3.5.0).
RNA sequencing and RamDA sequencing
RNA sequencing was performed as described in our previous study (Yokomizo-Nakano et al,
2020). RamDA sequencing was conducted as previously reported (Hayashi et al,
2018). Briefly, first-strand cDNA was synthesized from a 100-cell lysate using the PrimeScript RT reagent kit (TAKARA Bio) with 1st not-so-random (NSR) primers, and second-strand cDNA was then synthesized using Klenow Fragment (3′-5′ exo-; New England Biolabs) and 2nd NSR primers. Sequencing libraries were generated using the Nextera XT DNA Library Prep kit (Illumina) and sequenced on NextSeq500 (Illumina). Kallisto (v0.43.1) was used to calculate transcripts per million. R software was employed for statistical analyses and a k-means clustering analysis.
Quantitative RT-PCR (Q-RT-PCR)
Quantitative RT-PCR was performed on LightCycler 480 (Roche) using Luna Universal qPCR Master Mix (New England Biolabs). Expression levels were normalized to those of B2m. Primers for PCR were performed by using the following primers: Hmga2-Forward 5′-AGGCAGCAAAAACAAGAGCC-3′; Hmga2-Reverse 5′-CTGCCTCTTGGCCGTTTTTC-3′; B2m-Foward 5′-CTGGCTCACACTGAATTCACCCC-3′; B2m-Reverse 5′-TCGGCCATACTGGCATGCTTAAC-3′; Igf2bp2-Forward 5′-TCCCGGGTAGACATCCACAGAAA-3′; Igf2bp2-Reverse 5′-TCAGCCAGTTTGGTCTCATCAGC-3′; Hlf-Forward 5′-GCTTTGCCTTCTGCTCATCTGC-3′; Hlf-Reverse 5′-TGCTTTCTCACCTGCCTCCAAC-3′; Gfi1-Forward 5′-CAAACACTGATGCCCCCTGA-3′; Gfi1-Reverse 5′-ATCCCAAGTCAACCCTGCAA-3′; Tlr7-Forward 5′-CTGTTCTACTGGGGTCCAAAGCC-3′; Tlr7-Reverse 5′-TGGTTTCCATCCAGGTAAAGGGC-3′; Tlr8-Forward 5′-TGCCAAAGTCTGCTCTCTGCAC-3′; Tlr8-Reverse 5′-TGTTTTCCCCTTTCTGGCTGGG-3′; Ifi204-Forward 5′-CAGAAGTAACAGGAGAAACATCACT-3′; Ifi204-Reverse 5′-GTTGCAGAAGTCTCGCCTCT-3′.
Western blotting
Whole cell lysates were used for Western blotting. The following antibodies were used: FLAG (Sigma, M2 or Wako, 1E6), Hmga2 (CST, D1A7), HA (Abcam, ab9110), Phos-tag (Fujifilm, BTL-111), and Actin (Santa Cruz Biotechnology, C4).
Phosphoproteomic analysis
A phosphoproteomic analysis was performed by Proteobiologics (Osaka, Japan). Briefly, 1 × 10
7 BM MNCs were pooled from WT mice 3 days after the injection of 250 mg/kg 5-FU. A total of 200ug protein was boiled, reduced, and purified using SP3 protocol (Hughes et al,
2019). Purified protein was digested with trypsin and Lys-C. Desalted peptides were passed through the IMAC/C18 stage-tip, and phosphopeptides were purified on the lower C18 disc. The pooled TMT-labeled phosphopeptides were fractionated by off-line basic pH reversed-phase fractionation using Thermo Scientific UltiMate 3000 UHPLC system. Phosphopeptides were separated by L-column3 C18 column (Chemicals Evaluation and Research Institute), and the LC-MS/MS analysis was conducted by Q Exactive Plus (Thermo Fisher Scientific) coupled with Ultimate 3000 (Thermo Fisher Scientific) and HTC-PAL (CTC Analytics). Phosphopeptide identification and quantification were carried out with MaxQuant 1.6.14.0 supported by the Andromeda search engine, and the cut-off criterion for phosphosite probability was more than 0.75 (Sharma et al,
2014). The statistical analysis was carried out with Perseus 1.6.14.0 (Tyanova et al,
2016).
Statistical analysis
GraphPad Prism version 9 (GraphPad) was used to perform statistical analyses. The significance of differences was measured by an unpaired two-tailed Student’s t-test or the Mann–Whitney non-parametric test. A P value <0.05 was considered to be significant. No statistical methods were used to predetermine sample sizes for animal studies.
Acknowledgements
This work was supported in part by a grant from the Mochida Memorial Foundation for Medical and Pharmaceutical Research (to SK), the Takeda Science Foundation (to GS and MM), the Princess Takamatsu Cancer Research Fund (to GS), the Daiichi Sankyo Foundation of Life Science (to GS), the Japanese Society of Hematology (to GS), Grant-in-Aid for JSPS Fellows (22KJ2522 to MM), and Grants-in-Aid for Scientific Research (18H02842, 21K19512, 21H02952 (to GS), 21K08421 (to SK), 22K16304 (to JB), and 22H02904 (to MO)) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) of Japan.