Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2018, Vol. 12 Issue (5) : 559-565    https://doi.org/10.1007/s11684-017-0562-3
RESEARCH ARTICLE
Zfyve16 regulates the proliferation of B-lymphoid cells
Xuemei Zhao1, Donghe Li1, Qingsong Qiu1, Bo Jiao1, Ruihong Zhang1, Ping Liu1(), Ruibao Ren1,2()
1. State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Collaborative Innovation Center of System Biology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2. Department of Biology, Brandeis University, Waltham, MA 02454, USA
 Download: PDF(342 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Zfyve16 (a.k.a. endofin or endosome-associated FYVE-domain protein), a member of the FYVE-domain protein family, is involved in endosomal trafficking and in TGF-β, BMP, and EGFR signaling. The FYVE protein SARA regulates the TGF-β signaling pathway by recruiting Smad2/3 and accelerating their phosphorylation, thereby altering their susceptibility to TGF-β-mediated T cell suppression. Zfyve16 binds to Smad4 and their binding affects the formation of Smad2/3-Smad4 complex in TGF-β signaling. However, the in vivo function of Zfyve16 remains unknown. In this study, we generated a Zfyve16 knockout mouse strain (Zfyve16KO) and examined its hematopoietic phenotypes and hematopoietic reconstruction ability. The proportion of T cells in the peripheral blood of Zfyve16KO mice increases compared with that in wild-type mice. This finding is consistent with the role of Zfyve16 in facilitating TGF-β signaling. Unpredictably, B cell proliferation is inhibited in Zfyve16KO mice. The proliferation potential of Zfyve16KO B-lymphoid cells also significantly decreases in vitro. These results suggest that Zfyve16 inhibits the proliferation of T cells, possibly through the TGF-β signaling, but upregulates the proliferation of B-lymphoid cells.

Keywords Zfyve16      endofin      hematopoiesis      TGF-β      lymphocytes     
Corresponding Author(s): Ping Liu,Ruibao Ren   
Just Accepted Date: 30 October 2017   Online First Date: 18 December 2017    Issue Date: 29 September 2018
 Cite this article:   
Xuemei Zhao,Donghe Li,Qingsong Qiu, et al. Zfyve16 regulates the proliferation of B-lymphoid cells[J]. Front. Med., 2018, 12(5): 559-565.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-017-0562-3
https://academic.hep.com.cn/fmd/EN/Y2018/V12/I5/559
Fig.1  Targeted disruption of the Zfyve16 gene. (A) Allele architecture and identification of Zfyve16KO mice. Zfyve16 exons are indicated as gray boxes. FRT and loxP indicate recombinase recognition sites. (B) Zfyve16KO mice were genotyped by PCR analysis. (C) Representative semi-quantitative reverse-transcription PCR (SqRT-PCR) analysis was performed using RT-F and RT-R primers on exons VI and VII, respectively. RNA was extracted from bone marrow, spleen and liver. (D) Western blot analysis of Zfyve16 protein expression in kidney cells from Zfyve16KO and wild-type mice. (E) Body weights of Zfyve16KO and wild-type mice during the 7-month observation period.
Fig.2  Effect of Zfyve16 inactivation on peripheral blood cells. (A) Peripheral white blood cell (WBC), red blood cell (RBC), and platelet (PLT) counts of Zfyve16KO versus wild-type mice. (B) Percentage of B-lymphoid cells (CD19+ B220+), T-lymphoid cells (CD3e+), and myeloid cells (Gr-1+ Mac-1+) in the peripheral blood of Zfyve16KO and wild-type mice analyzed by flow cytometry. (C) Proportion of spleen B cells in each phase of the cell cycle. * P<0.05; ns, no statistical significance.
Fig.3  Effect of Zfyve16 depletion on hematopoietic organs and bone marrow cells. (A) Numbers of bone marrow cells and weights of spleen and liver of Zfyve16KO and wild-type mice. (B) Percentage of bone marrow hematopoietic stem cells (LSK), myeloid progenitor cells (GMP, CMP, and MEP), and committed lymphoid progenitors (CLP) in Zfyve16KO and wild-type mice. (C) Percentage of lineage cells in the bone marrow of Zfyve16KO and wild-type mice. * P<0.05; ns, no statistical significance.
Fig.4  Effect of Zfyve16 on the recovery of hematopoietic cells in response to irradiation. (A) Counts of WBC, RBC, and PLT in Zfyve16KO and wild-type mice following semi-lethal dose of irradiation. (B) Number of myeloid cells, T-lymphoid cells and B-lymphoid cells in Zfyve16KO and wild-type mice following semi-lethal dose of irradiation.
Fig.5  Colony-forming ability of bone marrow pre-B cells. (A) Representative photograph of colonies on day 10. (B) Quantitative comparison of colonies formed from the bone marrow of Zfyve16KO and wild-type mice. *** P<0.001.
1 Seet LF, Hong W. Endofin, an endosomal FYVE domain protein. J Biol Chem 2001; 276(45): 42445–42454
https://doi.org/10.1074/jbc.M105917200 pmid: 11546807
2 Michel SL, Berg JM. Building a metal binding domain, one half at a time. Chem Biol 2002; 9(6): 667–668
https://doi.org/10.1016/S1074-5521(02)00160-6 pmid: 12079775
3 Stenmark H, Aasland R, Toh BH, D’Arrigo A. Endosomal localization of the autoantigen EEA1 is mediated by a zinc-binding FYVE finger. J Biol Chem 1996; 271(39): 24048–24054
https://doi.org/10.1074/jbc.271.39.24048 pmid: 8798641
4 Stenmark H, Aasland R. FYVE-finger proteins—effectors of an inositol lipid. J Cell Sci 1999; 112(Pt 23): 4175–4183
pmid: 10564636
5 Gillooly DJ, Simonsen A, Stenmark H. Cellular functions of phosphatidylinositol 3-phosphate and FYVE domain proteins. Biochem J 2001; 355(2): 249–258
https://doi.org/10.1042/bj3550249 pmid: 11284710
6 Chen YG, Wang Z, Ma J, Zhang L, Lu Z. Endofin, a FYVE domain protein, interacts with Smad4 and facilitates transforming growth factor-b signaling. J Biol Chem 2007; 282(13): 9688–9695 PMID:17272273
https://doi.org/10.1074/jbc.M611704200
7 Dong M, Blobe GC. Role of transforming growth factor-b in hematologic malignancies. Blood 2006; 107(12): 4589–4596
https://doi.org/10.1182/blood-2005-10-4169 pmid: 16484590
8 Huang F, Chen YG. Regulation of TGF-b receptor activity. Cell Biosci 2012; 2(1): 9 doi:10.1186/2045-3701-2-9
pmid: 22420375
9 Toy W, Lim SK, Loh MC, Lim YP. EGF-induced tyrosine phosphorylation of endofin is dependent on PI3K activity and proper localization to endosomes. Cell Signal 2010; 22(3): 437–446
https://doi.org/10.1016/j.cellsig.2009.10.015 pmid: 19887107
10 Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor b in human disease. N Engl J Med 2000; 342(18): 1350–1358 doi:10.1056/NEJM200005043421807
pmid: 10793168
11 Khalil N. TGF-b: from latent to active. Microbes Infect 1999; 1(15): 1255–1263
https://doi.org/10.1016/S1286-4579(99)00259-2 pmid: 10611753
12 Hµger M, Pedersen CC, Larsen MT, Andersen MK, Hother C, Grønbæk K, Jarmer H, Borregaard N, Cowland JB. MicroRNA-130a-mediated down-regulation of Smad4 contributes to reduced sensitivity to TGF-b1 stimulation in granulocytic precursors. Blood 2011; 118(25): 6649–6659
https://doi.org/10.1182/blood-2011-03-339978 pmid: 22028478
13 Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-b family signalling. Nature 2003; 425(6958): 577–584
https://doi.org/10.1038/nature02006 pmid: 14534577
14 Letterio JJ. TGF-b signaling in T cells: roles in lymphoid and epithelial neoplasia. Oncogene 2005; 24(37): 5701–5712
https://doi.org/10.1038/sj.onc.1208922 pmid: 16123803
15 Zhang N, Bevan MJ. TGF-b signaling to T cells inhibits autoimmunity during lymphopenia-driven proliferation. Nat Immunol 2012; 13(7): 667–673
https://doi.org/10.1038/ni.2319 pmid: 22634866
16 Cottrez F, Groux H. Regulation of TGF-b response during T cell activation is modulated by IL-10. J Immunol 2001; 167(2): 773–778
https://doi.org/10.4049/jimmunol.167.2.773 pmid: 11441082
17 Letterio JJ, Geiser AG, Kulkarni AB, Dang H, Kong L, Nakabayashi T, Mackall CL, Gress RE, Roberts AB. Autoimmunity associated with TGF-b1-deficiency in mice is dependent on MHC class II antigen expression. J Clin Invest 1996; 98(9): 2109–2119
https://doi.org/10.1172/JCI119017 pmid: 8903331
18 Wolfraim LA, Fernandez TM, Mamura M, Fuller WL, Kumar R, Cole DE, Byfield S, Felici A, Flanders KC, Walz TM, Roberts AB, Aplan PD, Balis FM, Letterio JJ. Loss of Smad3 in acute T-cell lymphoblastic leukemia. N Engl J Med 2004; 351(6): 552–559
https://doi.org/10.1056/NEJMoa031197 pmid: 15295048
19 Bakkebø M, Huse K, Hilden VI, Smeland EB, Oksvold MP. TGF-b-induced growth inhibition in B-cell lymphoma correlates with Smad1/5 signalling and constitutively active p38 MAPK. BMC Immunol 2010; 11(1): 57
https://doi.org/10.1186/1471-2172-11-57 pmid: 21092277
20 Tsukazaki T, Chiang TA, Davison AF, Attisano L, Wrana JL. SARA, a FYVE domain protein that recruits Smad2 to the TGFb receptor. Cell 1998; 95(6): 779–791
https://doi.org/10.1016/S0092-8674(00)81701-8 pmid: 9865696
21 Itoh F, Divecha N, Brocks L, Oomen L, Janssen H, Calafat J, Itoh S, Dijke P. The FYVE domain in Smad anchor for receptor activation (SARA) is sufficient for localization of SARA in early endosomes and regulates TGF-b/Smad signalling. Genes Cells 2002; 7(3): 321–331
https://doi.org/10.1046/j.1365-2443.2002.00519.x pmid: 11918675
22 Seet LF, Hong W. Endofin recruits clathrin to early endosomes via TOM1. J Cell Sci 2005; 118(3): 575–587
https://doi.org/10.1242/jcs.01628 pmid: 15657082
23 Seet LF, Liu N, Hanson BJ, Hong W. Endofin recruits TOM1 to endosomes. J Biol Chem 2004; 279(6): 4670–4679
https://doi.org/10.1074/jbc.M311228200 pmid: 14613930
24 Goh JB, Wallace DF, Hong W, Subramaniam VN. Endofin, a novel BMP-SMAD regulator of the iron-regulatory hormone, hepcidin. Sci Rep 2015; 5(1): 13986
https://doi.org/10.1038/srep13986 pmid: 26358513
25 Shi W, Chang C, Nie S, Xie S, Wan M, Cao X. Endofin acts as a Smad anchor for receptor activation in BMP signaling. J Cell Sci 2007; 120(7): 1216–1224
https://doi.org/10.1242/jcs.03400 pmid: 17356069
26 Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-b1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 2007; 26(5): 579–591
https://doi.org/10.1016/j.immuni.2007.03.014 pmid: 17481928
[1] FMD-17047-OF-RRB_suppl_1 Download
[1] Kohei Miyazono, Yoko Katsuno, Daizo Koinuma, Shogo Ehata, Masato Morikawa. Intracellular and extracellular TGF-β signaling in cancer: some recent topics[J]. Front. Med., 2018, 12(4): 387-411.
[2] Xiaofeng Shi, Rong Ba, Haiyan You, Qian Jiang, Jiansong Huang, Jianhua Mao, Lanxiu Han, Shuo Zhang, Qin Zhuang, Xianqiu Yu, Lixia Wang, Yun Wang, Dongya Li, Wei Zhu, Yong Zhang, Yan Zhu, Xiaodong Xi. A rare case of B-lymphoproliferative disorder with villous lymphocytes harboring t(8;14)(q24;q32) translocation[J]. Front. Med., 2018, 12(3): 324-329.
[3] Joseph Cannova,Peter Breslin S.J.,Jiwang Zhang. Toll-like receptor signaling in hematopoietic homeostasis and the pathogenesis of hematologic diseases[J]. Front. Med., 2015, 9(3): 288-303.
[4] Xinguang Liu, Yu Hou, Jun Peng. Advances in immunopathogenesis of adult immune thrombocytopenia[J]. Front Med, 2013, 7(4): 418-424.
[5] Megan A. Hatlen, Lan Wang, Stephen D. Nimer. AML1-ETO driven acute leukemia: insights into pathogenesis and potential therapeutic approaches[J]. Front Med, 2012, 6(3): 248-262.
[6] Panpan Zhang, Feng Liu. In vivo imaging of hematopoietic stem cell development in the zebrafish[J]. Front Med, 2011, 5(3): 239-247.
[7] WU Kui, BI Yutian, WANG Yaoli, WANG Changzheng. Changes of phenotype and function of human CD4 CD25 T cells induced by transfection of Foxp3[J]. Front. Med., 2008, 2(4): 366-369.
[8] LU Ling, ZHANG Feng, PU Liyong, YAO Aihua, YU Yue, SUN Beicheng, LI Guoqiang, WANG Xuehao. Biological features of intrahepatic CD4+CD25+ T cells in the naturally tolerance of rat liver transplantation[J]. Front. Med., 2007, 1(4): 373-376.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed