Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration

Bi-Sen Ding, Daniel J Nolan, Jason M Butler, Daylon James, Alexander O Babazadeh, Zev Rosenwaks, Vivek Mittal, Hideki Kobayashi, Koji Shido, David Lyden, Thomas N Sato, Sina Y Rabbany, Shahin Rafii, Bi-Sen Ding, Daniel J Nolan, Jason M Butler, Daylon James, Alexander O Babazadeh, Zev Rosenwaks, Vivek Mittal, Hideki Kobayashi, Koji Shido, David Lyden, Thomas N Sato, Sina Y Rabbany, Shahin Rafii

Abstract

During embryogenesis, endothelial cells induce organogenesis before the development of circulation. These findings suggest that endothelial cells not only form passive conduits to deliver nutrients and oxygen, but also establish an instructive vascular niche, which through elaboration of paracrine trophogens stimulates organ regeneration, in a manner similar to endothelial-cell-derived angiocrine factors that support haematopoiesis. However, the precise mechanism by which tissue-specific subsets of endothelial cells promote organogenesis in adults is unknown. Here we demonstrate that liver sinusoidal endothelial cells (LSECs) constitute a unique population of phenotypically and functionally defined VEGFR3(+)CD34(-)VEGFR2(+)VE-cadherin(+)FactorVIII(+)CD45(-) endothelial cells, which through the release of angiocrine trophogens initiate and sustain liver regeneration induced by 70% partial hepatectomy. After partial hepatectomy, residual liver vasculature remains intact without experiencing hypoxia or structural damage, which allows study of physiological liver regeneration. Using this model, we show that inducible genetic ablation of vascular endothelial growth factor (VEGF)-A receptor-2 (VEGFR2) in the LSECs impairs the initial burst of hepatocyte proliferation (days 1-3 after partial hepatectomy) and subsequent reconstitution of the hepatovascular mass (days 4-8 after partial hepatectomy) by inhibiting upregulation of the endothelial-cell-specific transcription factor Id1. Accordingly, Id1-deficient mice also manifest defects throughout liver regeneration, owing to diminished expression of LSEC-derived angiocrine factors, including hepatocyte growth factor (HGF) and Wnt2. Notably, in in vitro co-cultures, VEGFR2-Id1 activation in LSECs stimulates hepatocyte proliferation. Indeed, intrasplenic transplantation of Id1(+/+) or Id1(-/-) LSECs transduced with Wnt2 and HGF (Id1(-/-)Wnt2(+)HGF(+) LSECs) re-establishes an inductive vascular niche in the liver sinusoids of the Id1(-/-) mice, initiating and restoring hepatovascular regeneration. Therefore, in the early phases of physiological liver regeneration, VEGFR2-Id1-mediated inductive angiogenesis in LSECs through release of angiocrine factors Wnt2 and HGF provokes hepatic proliferation. Subsequently, VEGFR2-Id1-dependent proliferative angiogenesis reconstitutes liver mass. Therapeutic co-transplantation of inductive VEGFR2(+)Id1(+)Wnt2(+)HGF(+) LSECs with hepatocytes provides an effective strategy to achieve durable liver regeneration.

Figures

Figure 1. Phenotypic signature and contribution of…
Figure 1. Phenotypic signature and contribution of LSECs to physiological liver regeneration induced by 70% partial hepatectomy (PH)
a) Liver sections obtained from VEGFR2-GFP reporter mice. During liver regeneration VEGFR2 is exclusively expressed on the liver ECs. b) Restricted expression of VEGFR3 on LSECs, but not CD34+ large vessels or hepatocytes. c) Polyvariate flow cytometric analysis of the liver nonparenchymal cells. VEGFR2+ cells that are CD45−, express EC-specific VE-cadherin. d) Specific expression of VEGFR3 on VEGFR2+VE-cadherin+CD45− LSECs, with a predominant fraction being CD34−FactorVIII+Prox-1−. Thus, LSECs could be identified as VEGFR3+CD34− cells. e) 48 hours after PH, E-cadherin+P-H3+ mitotic hepatocytes are localized adjacent to VE-cadherin+ and VEGFR2+ ECs. f, g) Kinetics of LSECs expansion (f) and hepatocyte mitosis (g) during liver regeneration (n = 4). Hpf, high power field. Scale bars, 50 μm. Error bars, s.e.m.
Figure 2. VEGFR2-Id1 activation in LSECs mediates…
Figure 2. VEGFR2-Id1 activation in LSECs mediates PH-induced liver regeneration
a, b) Hepatocyte proliferation after PH is impaired in VEGFR2fl/fl mice (n = 5). c-e) Inhibition of liver mass regeneration (c) and functional VE-cadherin+isolectin+ vessel formation (d, e) in VEGFR2fl/fl mice after PH (n = 4-6). f, g) Injection of VEGF-A164, but not VEGFR1-specific ligand PlGF, accelerates the regeneration of liver mass (f), associated with an incremental increase in VEGFR3+CD34− LSEC number (g) (n = 4). h) Regenerative liver section of Id1VenusYFP mouse. Id1 is selectively upregulated by PH in VE-cadherin+ vessels. i) VEGFR2 deletion diminishes Id1 upregulation in the regenerative liver (n = 5). *P < 0.05; #P < 0.01, versus VEGFR2fl/+ (b-e, i), versus PlGF-treated group (f). Scale bar, 50 μm. Error bars, s.e.m.
Figure 3. Id1 upregulation in LSECs is…
Figure 3. Id1 upregulation in LSECs is essential for liver regeneration
a) Compared to their wild type littermates (WT), Id1−/− mice manifest impaired regeneration in liver mass, which fails to be rescued by VEGF-A164 administration (n = 5). b, c) Impaired hepatocyte proliferation (b) and assembly of VE-cadherin+isolectin+ vessels (c) in the Id1−/− mice after PH (n = 5). d, e) The LSEC-dependent stimulation of hepatocyte proliferation was specifically inhibited by Id1 gene knockdown. Scr, scrambled. CM, LSEC-conditioned medium (n = 4). f) Intrasplenic transplantation of GFP-marked LSECs incorporates into the lumen of VEGFR3+ sinusoidal vasculature in the Id1−/− liver. g, h) Transplantation of Id1+/+ LSECs restores the regeneration of mass (g) and hepatocyte proliferation (h) in the Id1−/− liver (n = 4). Dashed line, level of Id1−/− liver without EC transplantation. k) Cellular proximity is essential in the stimulation of hepatocyte mitosis by the transplanted GFP+Id1+/+ vasculature. *P < 0.05, versus Id1−/− (a); #P < 0.01, versus Id1−/− with VEGF164 (a), versus WT (b, c). Scale bars, 50 (d, f) and 20 (h) μm. Error bars, s.e.m.
Figure 4. Id1-mediated induction of Wnt2 and…
Figure 4. Id1-mediated induction of Wnt2 and HGF in LSECs stimulates hepatic regeneration
a) Upregulation of HGF and Wnt2 is impaired in Id1−/− LSECs after PH (n = 5). b) Intrasplenic transplantation of GFP-marked Id1−/− LSECs carrying both Wnt2 and HGF (Id1−/−Wnt2+HGF+GFP+) rescues the regeneration of Id1−/− liver mass (n = 4). c) Transplantation of Id1−/−Wnt2+HGF+ LSECs restores the impaired hepatocyte proliferation in the Id1−/− liver. (n = 4). d) The proximity between the mitotic hepatocytes and the Id1−/−Wnt2+HGF+GFP+ LSECs in the Id1−/− liver. e) Requirement for VEGFR2-Id1 pathway in LSEC-mediated liver regeneration. Intrasplenic transplantation of Id1+/+ LSECs into the Id1−/− liver sinusoids restores hepatic-vascular regeneration. Transplanted Id1+/+ or Id1−/−Wnt2+HGF+GFP+ LSECs localize to the vicinity of hepatocytes, promoting inductive and proliferative angiogenesis thereby sustaining physiological liver regeneration. *P < 0.05; #P < 0.01. Scale bar, 20 μm. Error bars, s.e.m.

References

    1. Matsumoto K, Yoshitomi H, Rossant J, Zaret KS. Liver organogenesis promoted by endothelial cells prior to vascular function. Science. 2001;294:559–563.
    1. Lammert E, Cleaver O, Melton D. Induction of pancreatic differentiation by signals from blood vessels. Science. 2001;294:564–567.
    1. Sakaguchi TF, Sadler KC, Crosnier C, Stainier DY. Endothelial signals modulate hepatocyte apicobasal polarization in zebrafish. Curr. Biol. 2008;18:1565–1571.
    1. Makita T, Sucov HM, Gariepy CE, Yanagisawa M, Ginty DD. Endothelins are vascular-derived axonal guidance cues for developing sympathetic neurons. Nature. 2008;452:759–763.
    1. Butler JN, Vertes J, Varnum-Finney E, Kobayashi B, Hooper H, Seandel A, Shido M, White K, Kobayashi I, Witte M, May L, Shawber C, Kimura C, Kitajewski Y, Rosenwaks J, Bernstein Z, Rafii I, Endothelial S. Cells Are Essential for the Self-Renewal and Repopulation of Notch-Dependent Hematopoietic Stem Cells. Cell Stem Cell. 2010;6:1–14.
    1. Hooper AT, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell. 2009;4:263–274.
    1. Butler JM, Kobayashi H, Rafii S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer. 2010;10:138–146.
    1. McDonald B, et al. Interaction of CD44 and hyaluronan is the dominant mechanism for neutrophil sequestration in inflamed liver sinusoids. J. Exp. Med. 2008;205:915–927.
    1. Lee JS, Semela D, Iredale J, Shah VH. Sinusoidal remodeling and angiogenesis: a new function for the liver-specific pericyte? Hepatology. 2007;45:817–825.
    1. Klein D, et al. Wnt2 acts as a cell type-specific, autocrine growth factor in rat hepatic sinusoidal endothelial cells cross-stimulating the VEGF pathway. Hepatology. 2008;47:1018–1031.
    1. Greene AK, et al. Endothelial-directed hepatic regeneration after partial hepatectomy. Ann. Surg. 2003;237:530–535.
    1. Van Buren G, 2nd, et al. Effect of molecular therapeutics on liver regeneration in a murine model. J. Clin. Oncol. 2008;26:1836–1842.
    1. LeCouter J, et al. Angiogenesis-independent endothelial protection of liver: role of VEGFR-1. Science. 2003;299:890–893.
    1. Zaret KS, Grompe M. Generation and regeneration of cells of the liver and pancreas. Science. 2008;322:1490–1494.
    1. Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology. 2006;43:S45–53.
    1. Michalopoulos GK, DeFrances MC. Liver regeneration. Science. 1997;276:60–66.
    1. Greenbaum LE, et al. CCAAT enhancer- binding protein beta is required for normal hepatocyte proliferation in mice after partial hepatectomy. J. Clin. Invest. 1998;102:996–1007.
    1. Huh CG, et al. Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proc. Natl. Acad. Sci. U S A. 2004;101:4477–4482.
    1. Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol. Rev. 2008;88:125–172.
    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat. Med. 2003;9:669–676.
    1. Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438:932–936.
    1. Alitalo K, Tammela T, Petrova TV. Lymphangiogenesis in development and human disease. Nature. 2005;438:946–953.
    1. Lyden D, et al. Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts. Nature. 1999;401:670–677.
    1. Nam HS, Benezra R. High levels of Id1 expression define B1 type adult neural stem cells. Cell stem cell. 2009;5:515–526.
    1. Follenzi A, et al. Transplanted endothelial cells repopulate the liver endothelium and correct the phenotype of hemophilia A mice. J. Clin. Invest. 2008;118:935–945.
    1. Goessling W, et al. Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell. 2009;136:1136–1147.
    1. Ober EA, Verkade H, Field HA, Stainier DY. Mesodermal Wnt2b signalling positively regulates liver specification. Nature. 2006;442:688–691.
    1. Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease. Hepatology. 2007;45:1298–1305.
    1. Rafii S, Lyden D. Cancer. A few to flip the angiogenic switch. Science. 2008;319:163–164.
    1. Jin DK, et al. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nat. Med. 2006;12:557–567.

Source: PubMed

3
S'abonner