Human samples
This study included 18 healthy donors (20–68 years old, mean = 40; 11 females and 7 males), and 21 JAK2V617F-mutated patients (16–65 years old, mean = 37; 11 females and 10 males) from the Second Affiliated Hospital of Zhejiang University School of Medicine. Informed consent was obtained from all subjects. The study was approved by the Second Affiliated Hospital of Zhejiang University School of Medicine (No. 20230705). Blood samples were collected and the protein levels of TF and TFPI in plasma were measured by ELISA kit according to the manufacturer's instructions (R&D Systems, Minneapolis, MN, USA).
Animals
C57BL/6 mice were purchased from Zhejiang Provincial Laboratory Animal Center. EpoR-Cre mice were kindly provided by Stuart H. Orkin (Harvard Medical School, Boston, MA)55. LysM-Cre (Stock# 004781) and Jak2V617F/+ (Stock# 301658) mice were purchased from The Jackson Laboratory. Vav-iCre (Stock# C001019) and TFPIf/f (Stock# S-CKO-06215) mice were purchased from Cyagen Biosciences Inc. (Suzhou, China). CD169-Cre (Stock# NM-KI-215032) and Thbdf/f (Stock# NM-CKO-2101896) mice were obtained from Shanghai Model Organisms Center, Inc. (Shanghai, China). To generate erythroid lineage-specific and macrophage-specific TFPI knockout mice, EpoR-Cre and CD169-Cre mice were crossed with TFPIf/f mice on a C57BL/6 background. LysM-Cre mice were crossed with Thbdf/f mice to generate macrophage-specific Thbd knockout mice. CD169-DTR heterozygous (CD169DTR/+) mice on a C57BL/6 background56, which were generated with DTR complementary DNA (cDNA)57, were bred in house by crossing CD169DTR/DTR mice with C57BL/6 mice. Vav-iCre mice were crossed with Jak2V617F/+ mice to generate Vav-iCre;Jak2V617F/+ mice (Jak2V617F). All mice were housed in a specific pathogen-free barrier facility. Experiments were performed on 6–8-week-old mice. The experimental conditions and procedures were approved by the Zhejiang University Institutional Animal Care and Use Committee and were consistent with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.
Fech gene contains 11 exons, and exon 6 was selected as a conditional knockout region. PCR-generated homology arm and conditional knockout region were used to design targeting vector. Cas9, gRNA and targeting vector were co-injected into zygotes for the generation of Fechf/f mice. Mouse pups were genotyped by PCR and verified by sequencing. CD169-Cre mice were crossed with Fechf/f mice to generate macrophage-specific Fech knockout mice.
Reagents
Recombinant mouse TFPI proteins (mouse rTFPI; R&D Systems) were injected intravenously (i.v.) into mice at a dose of 50 µg/kg for 5 days. mouse rTFPI and human rTFPI (R&D Systems) were added to the cell culture medium at a concentration of 200 ng/ml. Anti-TFPI monoclonal antibodies were injected i.v. into mice at a dose of 5 mg/kg. SCH772984, a ERK1/2 inhibitor, was injected intraperitoneally (i.p.) into mice at a dose of 10 mg/kg for 7 days. AS1842856, a FOXO1 inhibitor, was injected i.p. into mice at a dose of 10 mg/kg for 7 days. JNK-IN-8, a JNK inhibitor, was injected i.p. into mice at a dose of 10 mg/kg for 7 days. In BrdU incorporation assays, 1 mg of BrdU was administered to mice by i.p. injection, Ter119+ CD71+ cells were collected and processed according to the manufacturer’s instructions in the BrdU Kit (BD Biosciences, San Jose, CA, USA).
Complete blood count analysis
Mice were bled to collect ~ 25 µl via the tail vein to collect blood in EDTA-coated BD Microtainer Blood Collection Tubes (BD, San Jose, CA, USA). Blood was diluted 1:20 in PBS and complete blood counts were measured on an Automatic Blood Analyzer (Sysmex, Kobe, Japan).
Real-time quantitative polymerase chain reaction (RT-qPCR)
Total RNA from tissue and cell samples were isolated using RNAiso reagent (TaKaRa, Dalian, China). After treatment with DNase I (Roche, Basel, Switzerland), reverse transcription was performed using AMV reverse transcriptase (TaKaRa) to obtain cDNA. Primers are listed in the Table S2. 18S rRNA was used as an internal reference gene. RT-qPCR was performed using an ABI StepOne Real-Time PCR System (Applied Biosystems, Foster City, CA) with TB Green Premix Ex Taq II (TaKaRa).
Western blot and co-immunoprecipitation (Co-IP) assays
For Western blot, tissue and cell samples were homogenized in lysis buffer (20 mM HEPES, 1.5 mM MgCl2, 0.2 mM EDTA, 100 mM NaCl, 0.2 mM dithiothreitol, 0.5 mM sodium orthovanadate, 0.4 mM PMSF, pH 7.4) containing phosphatase inhibitor (phosphatase inhibitor cocktail; Sigma-Aldrich). The soluble protein concentration was measured using the Bradford method. Proteins (20 µg of each sample) were separated by SDS-PAGE and electroporated onto polyvinylidene difluoride (PVDF) membranes. Then, non-fat milk was blocked, primary and secondary antibodies were incubated, and ECL reactions were performed.
For Co-IP, cells were washed with cold PBS and then lysed using lysis buffer for 1 h. The supernatants were collected after centrifugation 12,000 rpm for 10 min at 4°C and incubated with antibody-coupled Protein A beads or Protein G beads (Sigma-Aldrich) according to the manufacturer’s instructions. The beads were washed three times with lysis buffer, followed by Western blot analysis.
The following primary antibodies were used for Western blot. Anti-TFPI antibody (1:500, Abcam, Cambridge, UK), anti-Fech antibody (1:1000, Proteintech, Wuhan, China), anti-Thbd antibody (1:1000, Abcam, Cambridge, UK), anti-GATA1 antibody (1:1000, Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-p85 antibody (1:1000, Cell Signaling Technology, Boston, MA, USA), anti-Flag antibody (1:1000, Sigma-Aldrich), anti-Myc antibody (1:1000, Cell Signaling Technology), anti-GST antibody (1:1000, Cell Signaling Technology), anti-p-GATA1 antibody (1:500, Thermo Fisher Scientific, Waltham, MA, USA), anti-β-actin antibody (1:1000, Santa Cruz Biotechnology) and anti-GAPDH antibody (1:1000, Santa Cruz Biotechnology).
GST pull-down assay
GST pull-down was performed as described previously58. For tagging sequences with Flag, the cDNA encoding the sequence of TFPI was cloned into the pcDNA3.1 vector and then transfected into HEK293T cells in a 100-mm culture dish. GST-tagged Thbd protein was expressed in E. coli. BL21, the protein was purified and the GST-tagged Thbd protein was incubated with GSH-agarose in binding buffer (50 mM Tris/HCl, 150 mM NaCl, 1 mM EDTA, 0.5% NP40, 10% glycerol, pH 7.4), and rotated at 4°C for 1 h. Then, the beads loaded with the GST-tagged Thbd protein were collected and incubated with the Flag-tagged TFPI protein at 4°C for 2 h. The beads were washed 3 times followed by Western blot.
PHZ treatment
For induction of hemolytic anemia, mice were injected i.p. with PHZ (Sigma-Aldrich) at a dose of 40 mg/kg on days 0 and 1 of the experiment. Peripheral blood was collected 4 days before the start of treatment and on days 4, 7 and 12 after treatment.
Hypoxia exposure
Mice were exposed to hypoxia simulating an altitude of 5000 m (54.02 kPa, 10.8% O2) in a well-ventilated hypobaric chamber. Control mice were set at sea level (100.08 kPa, 20.9% O2) in the same chamber.
Flow cytometry and cell isolation
BM cells were isolated by thoroughly flushing tibias, femurs, and humeri using a 5 ml polystyrene tube with a strainer (BD Biosciences). Spleens were mashed through a 70 µm nylon filter. Cells were labeled with fluorochrome-conjugated antibodies in staining buffer for 30 min at 4°C. Samples were analyzed on a Gallios flow cytometer (Beckman Coulter, Miami, FL, USA). The analysis was performed using FlowJo software (Tree Star, Ashland, OR, USA).
Erythroid lineages were labeled with antibodies directed at CD71, Ter119 and CD44. Central macrophages were labeled with antibodies directed at Ter119, Ly6G, F4/80, VCAM-1, and CD169.
The following fluorescently labeled antibodies (BioLegend, San Diego, USA) were used: PE-anti-TER-119/Erythroid cells (clone Ter-119, 1:100), PE/Cyanine7-anti-CD71 (clone RI7217, 1:100), APC-anti-CD44 (clone IM7, 1:100), PE-anti-Ly6G (clone 1A8, 1:100), FITC-anti-TER-119/Erythroid cells (clone Ter-119, 1:100), BV421-anti-F4/80 (clone BM8, 1:25), and APC-anti-VCAM-1 (clone 429, 1:100). Flow analysis of live cells by exclusion of dead cells using propidium iodide (PI, Sigma-Aldrich). Identification of apoptotic cells were carried out using the FITC Annexin V Apoptosis Detection kit (BioLegend). For sorting of Lin− c-kit+ CD71+ cells, Ter119+ CD71+ cells, Ter119+ cells, erythroblasts and central macrophages, samples were processed under sterile conditions and sorted on FACS sorting with Moflo Astrios EQ (Beckman Coulter).
Colony-forming unit (CFU) assay
For CFU-E assay, the Lin− c-kit+ CD71+ cells were flow sorted and plated in erythropoietin-containing methylcellulose culture medium (StemCell Technologies, Vancouver, BC, Canada) and incubated at 37°C in 5% CO2 humidified atmosphere for 7 days. The number of colonies formed on each plate was counted using an inverted microscope.
Measurement of heme and Hb content
Intracellular heme content was determined according to fluorometric assays, as previously reported34. Briefly, cells were harvested and resuspended in 2 M oxalic acid and heated at 100°C for 30 min to remove iron from heme. The resultant protoporphyrin was measured by fluorescence (400 nm excitation and 662 nm emission). Endogenous protoporphyrin content was measured by detecting fluorescence in oxalic acid-treated unheated cells. The Hb content was quantified with the Drabkin’s reagent (Sigma-Aldrich).
Cell culture and transfection
HEK293T cells were obtained from American Type Culture Collection (ATCC) and were cultured in media composed of Dulbecco's Modified Eagle's Medium (DMEM), 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. Ter119+ CD71+ cells were sorted and cultured in media composed of Iscove's modified Dulbecco's medium (IMDM), 10% FBS, 1% bovine serum albumin, 0.2 mg/mL holotransferrin, 10 mg/mL insulin and erythropoietin at the different concentrations. Central macrophages were sorted and cultured in media composed of RPMI 1640, 10% FBS, 10 mM HEPES, and 10 ng/mL macrophage colony-stimulating factor (G-CSF). Cell viability was determined using trypan blue and counted with a hemocytometer. Cells were incubated at 37°C in 5% CO2 humidified atmosphere. HEK293T cells were transfected with pcDNA3.1 vector carrying the cDNA encoding sequence of TFPI, Thbd, and GATA1.
Macrophage depletion
To deplete CD169+ macrophages, heterozygous CD169DTR/+ mice were injected i.p. with diphtheria toxin (Sigma-Aldrich) at a dose of 10 µg/kg. In some experiments, macrophages were depleted by intravenous injection 200 µl of clodronate liposomes.
Lentivirus production and infection
For generation of mouse TFPI, aPC, CD71, GATA1, Thbd, TF, and human TFPI lentiviral vectors for knockdown, shRNA sequences targeting specific genes were synthesized and cloned into pLKO.1 vectors. The vectors were co-transfected into HEK293T cells with pSPAX2 (Addgene, Cambridge, MA, USA) and pMD2.G (Addgene, Cambridge, MA, USA) for packaging of lentiviral vectors. Lentiviral supernatants were collected 48 h post-transfection. For central macrophage infection, cells were transduced with lentivirus at a multiplicity of infection (MOI) of 10 and selected with puromycin (8 µg/ml) for 48 h. The knockdown efficiency was assessed by RT-qPCR and Western blot. Mice were injected with lentivirus at the dose of 6 × 108 pfu by tail vein injection. Target sequences are listed in Table S3.
Enzyme-linked immunosorbent assay (ELISA)
Femoral BM was rinsed with PBS and centrifuged to obtain cell supernatant. Blood samples were treated with sodium citrate and centrifuged at 4°C to extract plasma. The protein expression of TF and TFPI in plasma were measured by TF and TFPI ELISA kit according to the manufacturer's instructions (R&D Systems). The protein expression of TFPI-2 in BM supernatant or plasma were measured by TFPI-2 ELISA kit according to the manufacturer's instructions (USCN Life Science Inc., Wuhan, China).
In vitro human EBI formation
CD34+ cells were sorted by CD34 MicroBeads (Miltenyi Biotec, Germany) from human cord blood. Macrophages were derived from CD34+ cells by culturing in IMDM medium containing 2% human peripheral blood plasma, 3% human AB serum, 3 IU/mL heparin, 10 µg/mL insulin, 10 ng/mL stem cell factor (SCF), 1 ng/mL interleukin-3 (IL-3), 100 ng/mL macrophage colony-stimulating factor (M-CSF), 50 ng/mL fms-like tyrosine kinase 3 (FLT3), and 1 × penicillin-streptomycin. Erythroblasts were also derived from CD34+ cells. The cell culture procedure was comprised of 3 phases and 2 phased were used in present. In day 0 to day 6, CD34+ cells were cultured in IMDM containing 2% human peripheral blood plasma, 3% human AB serum, 200 µg/mL holo-human transferrin, 3 IU/mL heparin, 10 µg/ mL insulin, 10 ng/mL SCF, 1 ng/mL IL-3, and 3 IU/mL erythropoietin for 6 days. In day 7 to day 11, IL-3 was omitted from the culture medium. The Day 11 erythroblasts were pretreated with TFPI shRNA or control shRNA and mixed with macrophages at a 20:1 ratio. Then cells were cultured for 12 h in an IMDM medium containing 2% human peripheral blood plasma, 3% human AB serum, 3 IU/mL heparin, 10 µg/mL insulin, 200 µg/mL holo-human transferrin, 10 IU/ml EPO, 5 mM Mg2+, and 5 mM Ca2+. 1 × 105 cells were collected for cytospin analysis.
Transduction of JAK2 V617F mutation into CD34 + cells
The JAK2-transduced CD34+ cells were prepared as previously described59. Briefly, human wild type or mutant JAK2 cDNAs were respectively cloned into the MIGR1-IRES-GFP vector (Addgene, Cambridge, MA, USA). The vectors were co-transfected with lentivirus packaging plasmids pMD.G into HEK293T cells with Lipofectamine 3000. After 48 h, the lentiviral supernatants were collected, concentrated and stored at − 80°C. For infections, CD34+ cells were incubated with 50 µl of viral stock for 48 h.
RNA sequencing analysis
RNA was extracted from sorted central macrophages of TFPIf/f and TFPIf/f;EpoR mice, or BM cells of normoxia- and hypoxia-exposed mice. RNA quality was assessed by an Agilent 2100 Bioanalyzer (Agilent, Palo Alto, CA, USA) and quantified by a Nanodrop ND-2000 Spectrophotometer (Thermo Scientific, Waltham, MN, USA) prior to sequencing. High-quality RNA samples were used to construct sequencing libraries. RNA-seq transcriptome libraries were prepared using 1 µg of total RNA using the TruSeq RNA Sample Prep Kit from Illumina (San Diego, CA, USA). Libraries were sequenced using Illumina Novaseq 6000 with 2 × 151 bp read length. Expression levels for each transcript were using the fragments per kilobase of exon per million mapped reads (FPKM) method. Secreted proteins were identified as proteins carrying a signal peptide but lacking a transmembrane region60. Gene Oncology (GO) enrichment analysis were performed for the differentially expressed genes (DEGs) in the DAVID resource (https://david.ncifcrf.gov/). Kyoto Encyclopedia of genes and genomes (KEGG) path analysis of DEGs were carried out through clusterprofiler package in R. The ggplot2 package in R were used for Heatmap generation.
Quantification and statistical analysis
Data were shown as mean ± standard error of the mean (SEM). The biological repeats were indicated by ‘N’. Statistical analysis was performed using one-way ANOVA with SPSS (version 13.0) software. When the variances were significantly different (P < 0.05), logarithmic transformation was used to stabilize the variance. If the data did not have a normal distribution, statistical significance was evaluated using the Mann-Whitney U-test (two-tailed). P values < 0.05 was considered statistically significant.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.