GET THE APP

Progenitor Cells in Liver Development, Regeneration and Repair
Cell & Developmental Biology

Cell & Developmental Biology
Open Access

ISSN: 2168-9296

+44 1478 350008

Review Article - (2014) Volume 3, Issue 3

Progenitor Cells in Liver Development, Regeneration and Repair

Nirmala Mavila*, Marie V Nguyen, David James and Kasper S Wang
Developmental Biology and Regenerative Medicine and Stem Cell Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, USA
*Corresponding Author: Nirmala Mavila, Developmental Biology and Regenerative Medicine and Stem Cell Program, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, USA, Tel: +1 323-442-1100 Email:

Abstract

During organogenesis, the liver develops from the foregut endoderm and grows into the adjacent septum transversum resulting in the formation of the liver bud. Growth factors released from the septum transversum and the cardiac mesenchyme induce endodermal differentiation and proliferation, thus, forming the primordial liver and extrahepatic biliary structures. Endodermal precursor cells within the liver bud comprise bi-potential liver progenitor cells called hepatoblasts, which differentiates into hepatocytes and cholangiocytes. While the postnatal liver has remarkable capacity to regenerate via the proliferation of mature hepatocytes, this compensatory mechanism may be overwhelmed during states of chronic injury. Under these conditions, resident stem/progenitor cells proliferate to replace lost liver parenchyma. Significant progress has been made recently in elucidating the role of various signaling pathways and progenitor cells in liver development and regeneration. In this review, we summarize our recent understanding of progenitor cells in liver development, regeneration and repair.

<

Introduction

As the largest organ in the body, the liver is involved in numerous critical roles during homeostasis. The liver is organized into small hexagonal anatomic subunits, termed lobules, each consisting of a draining central vein and surrounded at their six corners by triads of hepatic portal veins, arteries, and bile ducts. Hepatocytes, which comprise over 85% of the total liver mass, reside alongside sinusoids, which carry admixed portal and arterial blood from the triads toward the draining central veins which themselves ultimately drain into hepatic veins. Hepatocytes are vital in terms of nutrient metabolism and storage, synthesis of secreted proteins, blood detoxification, and bile production amongst many key functions. Cholangiocytes or biliary epithelial cells, line the ducts that carry bile from the liver. Cholangiocytes are also involved in the absorption and secretion of water, lipids and electrolytes, thus maintaining ionic homeostasis of the bile. In addition, cholangiocytes play an important role in the local immune response by interacting with immune cells via secreted cytokines that in turn modulates the physiological functions of cholangiocytes as well as surrounding liver cells and invading inflammatory cells [1,2]. Hepatic stellate cells play a major role in liver regeneration following injury via secretion of growth factors such as Fibroblast Growth Factor (FGF), Hepatocyte Growth Factor (HGF), WNT (Wingless/Integrated), VEGF (Vascular Endothelial Growth Factor) and Insulin-like Growth Factor (IGF) [3,4]. Kupffer cells, the resident macrophages within the liver, clear aged red blood cells and pathogens present in the blood through phagocytosis. Sinusoidal endothelial cells line the capillaries in between the stacked hepatocytes within the lobule and play an important role in liver regeneration via production of growth factors such as HGF and EGF (Epidermal Growth Factor) [5].

Progenitor Cells in Liver Development

During early stages of hepatogenesis, a portion of the foregut endoderm invades the adjacent cardiac mesenchyme and septum transversum [6,7]. Both mesoderm-derived structures induce endodermal cells to proliferate, branch, and form the glandular epithelium of the liver via a combination of complex signaling pathways. Fibroblast Growth Factor (FGF), Bone Morphogenetic Protein (BMP) and Transforming Growth Factor-Beta (TGFβ) signaling pathways regulate liver bud induction during embryogenesis (Figure 1).

cell-developmental-liver-development

Figure 1: Cellular lineage during liver development

During this stage, hepatic endodermal cells undergo a morphologic transformation to epithelial cells, which attach to a basement membrane [7-14]. Cells within the liver bud express several hepatic genes including Albumin, Alpha-fetoprotein, and Transthyretin, along with several transcription factors, all of which are indicators of an early hepatic cell fate [7].

A subset of endodermal cells within the liver bud form a population of bi-potential progenitor cells, known as hepatoblasts, which express hepatocyte markers such as Albumin along with the biliary epithelial marker Cytokeratin-19 (Ck19) [7,8]. Additionally, putative stem/progenitor cell markers, such as Prominin-1 (also known as CD133) and CD49f, are expressed by murine hepatoblasts as early as embryonic day 12.5. FGF Receptor-2 (FGFR2)-mediated activation of AKT/β-catenin signaling via CREB binding protein (CBP) plays a critical role in the proliferation and survival of hepatoblasts during development [13,15].

During later stages of development, hepatoblasts differentiate into CK19-expressing biliary epithelial cells or HNF (Hepatocyte Nuclear Factor)-4α and Albumin-expressing hepatocytes. Gradients of growth factor signaling involving FGF, HGF (Hepatocyte Growth Factor) and TGFβ, BMP4, WNT (Wingless/Integrated) and Notch signaling pathways determine the fate of these cells toward either hepatocytes or biliary epithelial cells and are later organized into unique structures with distinct functions [10-12,16-21]. Portal fibroblasts regulate the differentiation of hepatoblasts towards a cholangiocyte phenotype and promote intrahepatic bile duct formation via Notch signaling [18-23]. Hepatoblasts at a greater distance from portal fibroblasts are thought to differentiate into hepatocytes and arrange into sheets of cells lined by sinusoidal epithelial cells and bile canaliculi. Several transcription factors such as GATA4, Hhex, HNFβ, HNF4α, HNF6, Tbox transcription factor 3, and Prox1 are known to regulate the hepatic specification of endodermal cells and later their differentiation to cholangiocytes or hepatocytes [14,24-30].

Liver Regeneration and Progenitor Cell Activation

The liver has a remarkable regenerative capacity compared to other organs. Mature hepatocytes have the ability to replicate and repopulate the liver during acute injury. Mesenchymal cells are known to have a critical role in liver regeneration. Stellate cells, fibroblasts, endothelial and kupffer cells are the main types of mesenchymal cells in the liver as well as the sources of growth factors during liver regeneration [3-5]. Following acute injury, hepatocytes release mitogenic signals to mesenchymal cells and induce their activation and proliferation. Activated mesenchymal cells in turn release growth factors and cytokines such as HGF, EGF, FGF, TNFα and IL6, all of which facilitate hepatocyte proliferation [31,32]. Under normal conditions, hepatocyte proliferation ceases one week after injury and liver mass is restored within two to three weeks. Studies suggest that TGFβ signaling may play an important role in this process [32-34] (Figure 2). However, the definitive molecular mechanism(s) that regulate the termination of hepatocyte proliferation during normal liver regeneration is unclear.

cell-developmental-chronic-injury

Figure 2: Liver regeneration during normal and chronic injury

During chronic injury, hepatocytes undergo ‘cellular senescence’, losing their proliferative capacity through the expression of cell cycle arrest proteins [35,36]. Studies demonstrate that Hepatic Progenitor Cells (HPCs), or oval cells, which are quiescent in the healthy liver, may be activated when the regenerative capacity of mature hepatocytes is severely impaired [37-39]. These activated postnatal HPCs express a variety of stem/progenitor markers such as EpCAM, OV6, Prominin1, CD49f, and A6 [15,40-43].

It is theorized that a stem cell niche populated by cholangiocytes, small hepatocytes, portal fibroblasts, and HPCs exists within the Canals of Hering, adjacent to the periportal regions of the liver [44]. When the liver is damaged, HPCs within this niche may become activated and differentiate into either cholangiocytes or hepatocytes (Figure 2). Intrinsic signals from the hepatic microenvironment regulate the expansion and differentiation of liver progenitor cells. FGF-mediated AKT/β-catenin signaling, for instance, modulates the presence of cells expressing the putative progenitor cell marker A6 in an experimental model of acute liver injury [45].

Whether this represents reprogramming of hepatocytes to express a progenitor marker or maturation and differentiation of progenitors toward a hepatocyte cell fate is unclear. There are lineage-tracing studies that provide evidences for hepatocyte differentiation of HPCs during injury. Recent study by utilizing a transgenic mouse model expressing inducible Cre recombinase under the control of Osteopontin gene, a known progenitor cell marker (46); demonstrated that HPCs contribute to hepatocyte mass in a choline deficient ethionine diet induced liver injury model (47). Another recent cell lineage study demonstrated that SRY-related HMG Box Transcription Factor-9 (SOX9) expressing cholangiocytes/progenitor cells are the source of hepatocytes in different experimental models of liver injury [48]. Upon transient amplification, these HPCs infiltrate along the liver plate toward the central vein and differentiate into hepatocytes to restore liver function and cell mass [47,48]. Despite all of these studies, the origin of progenitor cells and their migration during liver regeneration remains unclear [3-5]. Fibroblast progenitor cells are known to contribute to liver regeneration via activation of Hedghog signaling [49]. Macrophage-derived WNT is one of several factors, which influence the differentiation of progenitor cells towards hepatocyte lineage during liver regeneration [50]. Activated Notch signaling has been demonstrated during liver regeneration [51]. Given the known role of Notch signaling in biliary differentiation during liver development [18], it is plausible that this pathway also regulates progenitor cell differentiation into cholangiocytes during liver regeneration. Hence, cell-to-cell interactions and the liver microenvironment may influence the progenitor cell fate and thereby the extent of tissue regeneration during chronic liver injury.

Conclusions

Cellular homeostasis is a critical factor that determines proper organ development and regeneration. This is mainly achieved through a tight regulation of progenitor/stem cell proliferation and differentiation via a complex network of signaling pathways. Deregulation of these factors could lead to impaired development and tissue regeneration. Hence, a better understanding of the mechanisms by which liver progenitor cells interact with other liver cell types as well as the regulatory signaling pathways that get activated in response to injury is critical before implementing any cell based therapies for liver diseases.

Acknowledgements

MVN is supported by California Institute for Regenerative Medicine (CIRM) postdoctoral training grant.

References

  1. Fava G, Glaser S, Francis H, Alpini G (2005) The immunophysiology of biliary epithelium.Semin Liver Dis 25: 251-264.
  2. Strazzabosco M, Fabris L, Spirli C (2005) Pathophysiology of cholangiopathies.J ClinGastroenterol 39: S90-90S102.
  3. Yin C, Evason KJ, Asahina K, Stainier DY (2013) Hepatic stellate cells in liver development, regeneration, and cancer.J Clin Invest 123: 1902-1910.
  4. Friedman SL (2008) Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol 88:125-172.
  5. DeLeve LD (2013) Liver sinusoidal endothelial cells and liver regeneration.J Clin Invest 123: 1861-1866.
  6. Tremblay KD, Zaret KS (2005) Distinct populations of endoderm cells converge to generate the embryonic liver bud and ventral foregut tissues.DevBiol 280: 87-99.
  7. Zaret KS (2000) Liver specification and early morphogenesis.MechDev 92: 83-88.
  8. Zaret KS (2001) Hepatocyte differentiation: from the endoderm and beyond.CurrOpin Genet Dev 11: 568-574.
  9. Shin D, Shin CH, Tucker J, Ober EA, Rentzsch F, et al. (2007) Bmp and Fgf signaling are essential for liver specification in zebrafish.Development 134: 2041-2050.
  10. Calmont A, Wandzioch E, Tremblay KD, Minowada G, Kaestner KH, et al. (2006) An FGF response pathway that mediates hepatic gene induction in embryonic endoderm cells.Dev Cell 11: 339-348.
  11. Jung J, Zheng M, Goldfarb M, Zaret KS (1999) Initiation of mammalian liver development from endoderm by fibroblast growth factors.Science 284: 1998-2003.
  12. Rossi JM, Dunn NR, Hogan BL, Zaret KS (2001) Distinct mesodermal signals, including BMPs from the septum transversum mesenchyme, are required in combination for hepatogenesis from the endoderm.Genes Dev 15: 1998-2009.
  13. Berg T, Rountree CB, Lee L, Estrada J, Sala FG,et al. (2007) Fibroblast growth factor 10 is critical for liver growth during embryogenesis and controls hepatoblast survival via beta-catenin activation. Hepatology 46:1187-1197.
  14. Bort R, Signore M, Tremblay K, Martinez Barbera JP, Zaret KS (2006) Hex homeobox gene controls the transition of the endoderm to a pseudostratified, cell emergent epithelium for liver bud development.DevBiol 290: 44-56.
  15. MavilaN, James D, Utley S, Nguyen Cu,OrlyCoblens,et al. (2012) Fibroblast growth factor receptor-mediated activation of AKT-beta-catenin-CBP pathway regulates survival and proliferation of murine hepatoblasts and hepatic tumor initiating stem cells. PLoS One 7:e50401.
  16. Clotman F, Jacquemin P, Plumb-Rudewiez N, Pierreux CE, Van der Smissen P, et al. (2005) Control of liver cell fate decision by a gradient of TGF beta signaling modulated by Onecut transcription factors.Genes Dev 19: 1849-1854.
  17. Takayama K, Kawabata K, Nagamoto Y, Inamura M, Ohashi K,et al. (2014) CCAAT/enhancer binding protein-mediated regulation of TGFbeta receptor 2 expression determines the hepatoblast fate decision. Development 141:91-100.
  18. Zong Y, Panikkar A, Xu J, Antoniou A, Raynaud P, et al. (2009) Notch signaling controls liver development by regulating biliary differentiation.Development 136: 1727-1739.
  19. Clotman F, Lemaigre FP (2006) Control of hepatic differentiation by activin/TGFbeta signaling.Cell Cycle 5: 168-171.
  20. Jeliazkova P, Jors S, Lee M, Zimber-Strobl U, Ferrer J,et al. (2013) Canonical Notch2 signaling determines biliary cell fates of embryonic hepatoblasts and adult hepatocytes independent of Hes1. Hepatology 57: 2469-2479.
  21. Ader T, Norel R, Levoci L, Rogler LE (2006) Transcriptional profiling implicates TGFbeta/BMP and Notch signaling pathways in ductular differentiation of fetal murine hepatoblasts.MechDev 123: 177-194.
  22. Kodama Y, Hijikata M, Kageyama R, Shimotohno K, Chiba T (2004) The role of notch signaling in the development of intrahepatic bile ducts.Gastroenterology 127: 1775-1786.
  23. Lemaigre FP (2003) Development of the biliary tract.MechDev 120: 81-87.
  24. Lee CS, Friedman JR, Fulmer JT, Kaestner KH (2005) The initiation of liver development is dependent on Foxa transcription factors.Nature 435: 944-947.
  25. Parviz F, Matullo C, Garrison WD, Savatski L, Adamson JW, et al. (2003) Hepatocyte nuclear factor 4alpha controls the development of a hepatic epithelium and liver morphogenesis.Nat Genet 34: 292-296.
  26. Bossard P, Zaret KS (1998) GATA transcription factors as potentiators of gut endoderm differentiation.Development 125: 4909-4917.
  27. Clotman F, Lannoy VJ, Reber M, Cereghini S, Cassiman D, et al. (2002) The onecut transcription factor HNF6 is required for normal development of the biliary tract.Development 129: 1819-1828.
  28. Zaret KS, Watts J, Xu J, Wandzioch E, Smale ST,et al. (2008) Pioneer factors, genetic competence, and inductive signaling: programming liver and pancreas progenitors from the endoderm. Cold Spring HarbSymp Quant Biol 73:119-126.
  29. Zaret KS, Grompe M (2008) Generation and regeneration of cells of the liver and pancreas.Science 322: 1490-1494.
  30. Wandzioch E, Zaret KS (2009) Dynamic signaling network for the specification of embryonic pancreas and liver progenitors.Science 324: 1707-1710.
  31. Böhm F, Köhler UA, Speicher T, Werner S (2010) Regulation of liver regeneration by growth factors and cytokines.EMBO Mol Med 2: 294-305.
  32. Fausto N, Laird AD, Webber EM (1995) Liver regeneration. 2. Role of growth factors and cytokines in hepatic regeneration.FASEB J 9: 1527-1536.
  33. Nishikawa Y, Wang M, Carr BI (1998) Changes in TGF-beta receptors of rat hepatocytes during primary culture and liver regeneration: increased expression of TGF-beta receptors associated with increased sensitivity to TGF-beta-mediated growth inhibition.J Cell Physiol 176: 612-623.
  34. Jirtle RL, Carr BI, Scott CD (1991) Modulation of insulin-like growth factor-II/mannose 6-phosphate receptors and transforming growth factor-beta 1 during liver regeneration.J BiolChem 266: 22444-22450.
  35. Michalopoulos GK (2007) Liver regeneration.J Cell Physiol 213: 286-300.
  36. Marshall A, Rushbrook S, Davies SE, Morris LS, Scott IS, et al. (2005) Relation between hepatocyte G1 arrest, impaired hepatic regeneration, and fibrosis in chronic hepatitis C virus infection.Gastroenterology 128: 33-42.
  37. Santoni-Rugiu E, Jelnes P, Thorgeirsson SS, Bisgaard HC (2005) Progenitor cells in liver regeneration: molecular responses controlling their activation and expansion.APMIS 113: 876-902.
  38. Theise ND, Saxena R, Portmann BC, Thung SN, Yee H, et al. (1999) The canals of Hering and hepatic stem cells in humans.Hepatology 30: 1425-1433.
  39. Itoh T, Miyajima A (2014) Liver regeneration by stem/progenitor cells.Hepatology 59: 1617-1626.
  40. Yoon SM, Gerasimidou D, Kuwahara R, Hytiroglou P, Yoo JE, et al. (2011) Epithelial cell adhesion molecule (EpCAM) marks hepatocytes newly derived from stem/progenitor cells in humans.Hepatology 53: 964-973.
  41. Roskams T, De Vos R, Van Eyken P, Myazaki H, Van Damme B, et al. (1998) Hepatic OV-6 expression in human liver disease and rat experiments: evidence for hepatic progenitor cells in man.J Hepatol 29: 455-463.
  42. Stamp LA, Braxton DR, Wu J, Akopian V, Hasegawa K, et al. (2012) The GCTM-5 epitope associated with the mucin-like glycoprotein FCGBP marks progenitor cells in tissues of endodermal origin.Stem Cells 30: 1999-2009.
  43. Petersen BE, Grossbard B, Hatch H, Pi L, Deng J, et al. (2003) Mouse A6-positive hepatic oval cells also express several hematopoietic stem cell markers.Hepatology 37: 632-640.
  44. Kuwahara R, Kofman AV, Landis CS, Swenson ES, Barendswaard E, et al. (2008) The hepatic stem cell niche: identification by label-retaining cell assay.Hepatology 47: 1994-2002.
  45. Utley S, James D, Mavila N, Nguyen MV, Vendryes C, et al. (2014) Fibroblast growth factor signaling regulates the expansion of A6-expressing hepatocytes in association with AKT-dependent β-catenin activation.J Hepatol 60: 1002-1009.
  46. Coombes JD, Swiderska-Syn M, Dollé L, Reid D, Eksteen B, et al. (2014) Osteopontinneutralisation abrogates the liver progenitor cell response and fibrogenesis in mice.Gut .
  47. Español-Suñer R, Carpentier R, Van Hul N, Legry V, Achouri Y, et al. (2012) Liver progenitor cells yield functional hepatocytes in response to chronic liver injury in mice.Gastroenterology 143: 1564-1575.
  48. Furuyama K, Kawaguchi Y, Akiyama H, Horiguchi M, Kodama S, et al. (2011) Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine.Nat Genet 43: 34-41.
  49. Swiderska-Syn M, Syn WK, Xie G, Krüger L, Machado MV, et al. (2014) Myofibroblastic cells function as progenitors to regenerate murine livers after partial hepatectomy.Gut 63: 1333-1344.
  50. Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, et al. (2012) Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease.Nat Med 18: 572-579.
  51. Köhler C, Bell AW, Bowen WC, Monga SP, Fleig W, et al. (2004) Expression of Notch-1 and its ligand Jagged-1 in rat liver during liver regeneration.Hepatology 39: 1056-1065.
Citation: Mavila N, Marie V N, James D and Wang S K (2014) Progenitor Cells in Liver Development, Regeneration and Repair. Cell Dev Biol 3:144.

Copyright: © 2014 Mavila N, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top