Introduction
The
Zfyve16 gene encodes endofin (endosome-associated FYVE-domain protein), an evolutionarily conserved large protein of 250 kDa [
1]. Endofin is a member of the FYVE domain family and contains a highly conserved FYVE motif and a Smad binding domain (SBD). The FYVE domain is a highly conserved zinc-finger domain and contains eight conserved cysteine residues, which coordinate two Zn
2+ ions in a “cross-braced” topology [
2–
4]. This domain also contains other conserved residues, such as the R(R/K)HHCRxCG motif surrounding a third and a fourth cysteine residue and arginine at position 74 [
5]. With its Zn
2+ coordination and a basic motif, the FYVE domain family proteins can bind to phosphatidylinositol-3-phosphate on the cytoplasmic surface of cell membranes [
6–
9]. The membrane association ability allows FYVE-containing proteins to recruit additional molecules to localize to the cell membrane.
The TGF-
b signaling pathway regulates many cellular processes, including cell proliferation, differentiation, and tumor metastasis. In the hematological system, TGF-
b negatively regulates cell proliferation and differentiation [
10,
11]. TGF-
b is initially secreted as a latent protein complex and requires activation to exert its biological function [
12,
13]. The activated TGF-
b signaling pathway is primarily mediated by two receptors, namely, type II TGF-
b receptor (T
bRII) and type I TGF-
b receptor (T
bRI), which are serine/threonine kinase receptors. The TGF-
b family ligands first bind to the T
bRII receptor. After its autophosphorylation, T
bRII phosphorylates and activates T
bRI, which then phosphorylates a series of downstream signaling molecules, including receptor-regulated Smads (R-Smads) [
14]. The recruitment and phosphorylation of R-Smads are key steps in the TGF-
b signaling pathway and play an important role in TGF-
b signaling.
The disruption of TGF-
b signaling in T cells impairs the maintenance of regulatory T cells, leading to the expansion of the activated effector T cells [
15–
18]. A previous mouse knockout experiment reported that Smad3 plays a critical role in mediating the inhibitory effects of TGF-
b signaling in T cells [
18]. TGF-
b signaling also inhibits the proliferation and differentiation of B lymphocytes. In particular, TGF-
b-induced Smad1/5 phosphorylation inhibits the proliferation of B lymphocytes [
19].
TGF-
b signaling pathways can be enhanced by scaffold proteins, such as SARA, which can recruit Smad2/3 to the membrane and accelerate their phosphorylation [
20]. Similar to Zfyve16, SARA contains an FYVE domain and an SBD domain. The FYVE domain in SARA can recruit Smad2/3 to the vicinity of the activated T
bRI and phosphorylate them [
21]. Phospho-Smad2/3 binds to the common Smad (Smad4) to form a complex, which transfers into nuclear regulatory downstream target genes [
6].
Similar to SARA, Zfyve16 is enriched in EEA1-positive endosomes and functions as a scaffold protein to facilitate TGF-
b signaling [
22,
23]. In TGF-
b signaling, Zfyve16 promotes Smad3 and Smad4 to form a heteromeric complex, which translocates to the nucleus and controls the transcription of target genes [
6]. The target genes include apoptosis regulators, namely, Bcl-xl, Bim, Bax, and cell cycle regulators such as DAPK1. In addition, Zfyve16 binds to Smad1, and the bonding enhances Smad1 phosphorylation and nuclear localization [
24]. Despite these biochemical studies, the
in vivo function of Zfyve16 remains unknown.
In this study, we generated a murine model with deletion of the Zfyve16 gene. The proportion of T cells in Zfyve16KO mice increases compared with that in the wild-type mice. However, Zfyve16 deficiency exhibits an opposite effect on B-lymphoid cells. Hence, Zfyve16 plays a novel role in B lymphocyte development.
Materials and methods
Zfyve16 knockout mice
Zfyve16-deficient mice containing a “knockout-first” allele targeted to the Zfyve16 genomic locus named Zfyve16tm2a(KOMP)Wtsi were obtained from the Knockout Mouse Project resource (KOMP-CSD ID:41673). Mice were genotyped using DNA extracted from mouse tails through multiplex PCR with the following primers (wild-type= 491 bp; knockout= 399 bp):
Zfyve16-F: GAGATGGCGCAACGCAATTAATG,
Zfyve16-R: TTAAGCTCAACTCAAGGCTTTCGCC,
Lox-F: GAGATGGCGCAACGCAATTAATG,
Lox-R: GTCACAGTAGCCCAAACTCACATGC.
Semi-quantitative PCR primer:
RT-F: GCCGAATGGTGAAGTTGCAG,
RT-R: TGACAAGTTG GACTCTGGCT.
Mice were bred and kept in barrier facilities. All procedures were carried out in accordance with the ethical approval of the Animal Care and Welfare Committee of Shanghai Jiao Tong University School of Medicine.
Western blot analysis
Mouse kidney cells were treated with red blood cell lysis buffer and lysed with 1× lysis buffer (#9803, Cell Signaling Technology, MA, USA) containing protease inhibitors and 1 mmol/L PMSF. Protein concentration in the supernatant was determined using a BCA Protein Assay Kit (Thermo Scientific, USA). Equal amounts of total protein from each group were separated on 10% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membrane was incubated with mouse polyclonal anti-endofin antibody (1:500, A7766, Abcam, Cambridge, UK) and anti-Gapdh antibody (1:1000; Cell Signaling Technology, MA, USA). Goat-anti-rabbit IgG was used as secondary antibody.
Flow cytometry analysis
For surface staining, cells isolated from mice were treated with red blood cell lysis buffer at room temperature for 5 min and washed in cold PBS. Cells (1 × 106 per assay) were incubated with conjugated antibodies (5 µg/mL) in the dark at room temperature for 20 min. Gr-1-PE, Mac-1-PE-cy7, B220-APC-cy7, and CD19-APC (BD, San Diego, CA, USA) were used to analyze hematopoietic cells of different lineages. Lineage-APC-cy7, Sca-1-PE-cy7, c-kit-APC, CD127-PE, CD16/32-Percp-cy5.5, and CD34-PE (BD, San Diego, CA, USA) were used to analyze hematopoietic stem cells (HSC) and progenitor cells (HSPCs). Nuclear staining of Ki67 was performed using FITC-conjugated Ki67 antibody (BD Pharmingen, CA, USA) and 1 × fixation/permeabilization solutions (BD Biosciences). After washing with cold PBS, the cells were resuspended in 200 µL of PBS and subjected to fluorescence-activated cell sorting (FACS) machine (LSR II system, BD Biosciences, San Jose, CA, USA). FACS data were analyzed using FlowJo, 7.6.1 software (Tree Star, San Carlos, CA, USA).
Blood cell counts
Briefly, 10 µL of blood collected from each mouse by tail vein bleeding was transferred into 1.5 mL Eppendorf tube containing 1 µL of 0.5 mol/L EDTA (BD, Pharmingen, CA, USA). Complete blood count (CBC) was determined using a pocH-100iV Diff hematology analyzer (Sysmex Corporation, Kobe, Japan).
Colony formation assay
Colony forming unit (CFU)-Pre-B assays were performed using MethoCult M3630 methylcellulose medium (StemCell Technologies, Vancouver, Canada) in accordance with the manufacturer’s instructions. Mouse bone marrow cells were isolated, plated into 3 mL of MethoCult M3630 methylcellulose medium at a density of 2 × 105 cells/dish in triplicate, and cultured at 37 °C and 5% CO2. Colonies were counted after 10 days of culture.
Statistical analysis
GraphPad Prism 5 software was used for statistical analysis of data. Statistical significance threshold was set at 0.05.
Results
Characterization of Zfyve16 knockout mice
Zfyve16-deficient mice were generated by homologous recombination in embryonic stem cells with the Zfyve16 gene locus targeted by a gene trapping cassette (Zfyve16tm2a(KOMP)Wtsi), which contains a splice acceptor site En2SA, an internal ribosome entry site IRES, and a lacZ reporter between Zfyve16 exons VI and VII (Fig. 1A). The Zfyve16 knockout mouse strain was confirmed by tail-DNA PCR analysis (Fig. 1B). The disruption of the expression of Zfyve16 transcripts by the trapping cassette was confirmed by RT-qPCR analysis, although exon 7 was slightly expressed in the knockout mice (Fig. 1C). Western blot analysis showed that Zfyve16 was not expressed at the protein level because the trapping cassette also caused a frameshift mutation in Zfyve16 mRNA (Fig. 1D).
Zfyve16 knockout mice (Zfyve16KO) were viable and fertile and exhibited no apparent abnormalities compared with the wild-type littermates. In addition, the body weight of Zfyve16 knockout mice was not significantly different from that of the wild-type mice during the 9-month observation period (Fig. 1E).
Zfyve16 modulates the proportions of lymphoid cells in peripheral blood
To examine the effect of Zfyve16 deficiency on hematopoiesis, we compared the peripheral blood of Zfyve16KO and wild-type mice. The numbers of white blood cells (WBCs), red blood cells (RBCs), and platelets in the peripheral blood of Zfyve16KO mice are similar to those in the wild-type controls (Fig. 2A). The frequency of myeloid cells (Gr-1+Mac-1+) in peripheral blood was not significantly different between Zfyve16KO and wild-type mice (Fig. 2B). However, the percentage of T-lymphoid cells (CD3e+) in the peripheral blood of Zfyve16KO mice is relatively higher than that in wild-type mice (Fig. 2B), while the proportion of B-lymphoid cells (CD19+ B220+) decreased compared with that of control mice. Hence, Zfyve16 regulates the development of lymphoid cells.
We also examined the proliferation of B cells in spleen, considering that most B cells in peripheral blood are quiescent. The number of splenic B cells in the G0 phase is significantly higher in Zfyve16KO mice compared with that in wild-type mice (Fig. 2C). We also analyzed the apoptosis of B-lymphoid cells from bone marrow, spleen, and peripheral blood by using annexin V and 7AAD. The proportion of apoptotic B cells was not significantly different between Zfyve16KO and Zfyve16WT mice (Supplementary Fig. 1). These data indicate that Zfyve16 regulates B cell proliferation by regulating the cell cycle.
Zfyve16 modulates the frequencies of common lymphoid progenitors
We further investigated the physiological role of Zfyve16 on hematopoietic stem and progenitor cells (HSPCs). The function of Zfyve16 in the bone marrow, spleen, liver, thymus, and lymph nodes was not significantly different between Zfyve16KO and wild-type mice (Fig. 3A and data not shown). Bone marrow analysis revealed no significant changes in LSK (Lin-Sca-1+c-Kit+) cells and in myeloid progenitor (CMP, Lin−Sca-1−c-Kit+FcgRhiCD34+), granulocytic and monocytic progenitor (GMP, Lin-Sca-1-c-Kit+FcgRhiCD34+), and megakaryocytic and erytheral progenitor (MEP, Lin−Sca-1−c-Kit+FcgRhiCD34+) cells between Zfyve16KO and wild-type mice (Fig. 3B). However, the number of committed lymphoid progenitor (CLP, Lin−c-kitloCD127+) cell subpopulation significantly increased in Zfyve16KO mice compared with that in wild-type mice. This phenomenon is consistent with the increased number of T-lymphoid cells in Zfyve16KO and wild-type mice.
Zfyve16 modulates the reconstitution abilities of T- and B-lymphoid cells in recipient mice
Considering the differential effect of Zfyve16 on T- and B-lymphoid cells in steady hematopoiesis, we investigated whether such difference exists during bone marrow reconstitution and in response to external stress. We conducted adoptive bone marrow transplantation experiment, in which Zfyve16KO (CD45.2)/Zfyve16WT (CD45.2) and wild-type (CD45.1) BM cells were mixed (1:1, 2 × 105 each) and transplanted into lethally irradiated recipient mice (CD45.1). The result of FACS analysis showed that the percentage of donor derived B cells was not significantly different between Zfyve16KOand Zfyve16WT mice (Supplementary Fig. 2). This finding demonstrates that the bone marrow reconstitution capacity is not affected by the loss of Zfyve16. In another separate experiment, Zfyve16KO and wild-type mice were subjected to semi-lethal dose of radiation. The recovery of the hematopoietic system was monitored every 3 days after irradiation (Fig. 4A and 4B). Blood cells began to recover on the 9th day after irradiation. The counts of WBC, RBC, and platelet in peripheral blood were not significantly different between the two groups of mice (Fig. 4A). Moreover, the course of recovery of myeloid cells was similar between the two groups. However, the recovery rate of B-lymphoid cells is significantly lower in Zfyve16KO mice compared with that in wild-type mice. These results suggest that Zfyve16 may regulate the proliferation of B cells. By contrast, the proliferation rate of T-lymphoid cells is significantly higher in Zfyve16KO mice than in wild-type mice. These data confirm that Zfyve16 affects the development of lymphoid lineage cells.
Zfyve16 modulates the colony formation capacity of B-lymphoid cells
TGF-
b signaling negatively regulates T cell expansion [
14,
15]. Similarly, the present results showed that the depletion of Zfyve16, a positive regulator of TGF-
b signaling, increased the number of T-lymphoid cells. However, the effect of Zfyve16 on B cells was not predicted. To further confirm the effect of Zfyve16 on B cell proliferation, we performed a B-lymphoid cell colony formation assay. Briefly, 2 × 10
5 bone marrow cells were plated in triplicate into the methylcellulose medium for culturing mouse B-lymphoid cells (MethoCult
TMM3630). Colonies were counted on day 10 after plating. Both the size and number of B cell colonies in
Zfyve16KO mice significantly decreased compared with those in wild-type mice (Fig. 5A and 5B). The data demonstrate that Zfyve16 directly regulates the growth of B-lymphoid cells.
Discussion
Zfyve16 plays a critical role in TGF-
b signal transduction and endosomal trafficking [
1,
25]. In this study, we investigated the function of Zfyve16
in vivo by using mice with inactivating insertion into the Zfyve16 alleles.
Zfyve16KO mice developed normally and showed no gross abnormalities. The proportion of T cells in the peripheral blood of
Zfyve16KO mice is significantly, though not dramatically, higher than that in wild-type mice. Consistently, the number of common lymphoid progenitor cells in
Zfyve16KO mice also increased. Zfyve16 forms complexes with Smad2/3 and Smad4 in TGF-
b signaling, and Smad2/3 complexes are involved in TGF-
b inhibition of interleukin-2 (IL-2), interferon-
g (IFN-
g) and granzyme B transcription, which are required for proliferation and differentiation of effector T helper 1 (Th1), Th2, and cytotoxic T lymphocyte cells [
14,
26]. The inactivation of Zfyve16 possibly weakens TGF-
b signaling and ameliorates the negative regulatory role of TGF-
b in T-lymphoid cell proliferation. The increased number of T-lymphoid cells in
Zfyve16KO mice is consistent with previous findings.
Unexpectedly, we found that the proportion of B-lymphoid cells in
Zfyve16KO mice decreased. The B-cell colony assay confirmed the
in vivo observation, indicating that this effect is cell autonomous. These results demonstrate for the first time that Zfyve16 positively regulates the development of B-lymphoid cells. The decreased proliferation of B-lymphoid cells in
Zfyve16KO mice is inconsistent with the negative regulatory role of TGF-
b signaling in lymphocyte development and activation [
19]. Hence, Zfyve16 possibly regulates other cellular processes which are required for the full proliferation capacity of B-lymphoid cells. According with the increasing proportion of CLP, the reduction in the number of B cells could be due to the blockade of CLP differentiation into B-lymphoid cells. Further studies must investigate such novel mechanisms in the future.
Higher Education Press and Springer-Verlag GmbH Germany, part of Springer Nature