SciELO - Scientific Electronic Library Online

 
vol.106 número3¿Es la coagulación con plasma argón una opción terapéutica eficaz y segura en pacientes con proctitis crónica actínica tras dosis altas de radioterapia?Sedación en endoscopia digestiva: guía de práctica clínica de la Sociedad Española de Endoscopia Digestiva índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Revista Española de Enfermedades Digestivas

versión impresa ISSN 1130-0108

Rev. esp. enferm. dig. vol.106 no.3 Madrid mar. 2014

 

REVIEW

 

Liver regeneration - The best kept secret. A model of tissue injury response

Regeneración hepática; el secreto mejor guardado. Una forma de respuesta al daño tisular

 

 

Javier A.-Cienfuegos, Fernando Rotellar, Jorge Baixauli, Fernando Martínez-Regueira, Fernando Pardo and José Luis Hernández-Lizoáin

Department of General Surgery. Clínica Universidad de Navarra. Pamplona, Navarra. Spain

Correspondence

 

 


ABSTRACT

Liver regeneration (LR) is one of the most amazing tissue injury response. Given its therapeutic significance has been deeply studied in the last decades.
LR is an extraordinary complex process, strictly regulated, which accomplishes the characteristics of the most evolutionary biologic systems (robustness) and explains the difficulties of reshaping it with therapeutic goals.
TH reproduces the physiological tissue damage response pattern, with a first phase of priming of the hepatocytes -cell-cycle transition G0-G1-, and a second phase of proliferation -cell-cycle S/M phases- which ends with the liver mass recovering.
This process has been related with the tissue injury response regulators as: complement system, platelets, inflammatory cytokines (TNF-α, IL-1β, IL-6), growth factors (HGF, EGF, VGF) and anti-inflammatory factors (IL-10, TGF-β).
Given its complexity and strict regulation, illustrates the unique alternative to liver failure is liver transplantation.
The recent induced pluripotential cells (iPS) description and the mesenchymal stem cell (CD133+) plastic capability have aroused new prospects in the cellular therapy field. Those works have assured the cooperation between mesenchymal and epithelial cells.
Herein, we review the physiologic mechanisms of liver regeneration.

Key words: Liver regeneration. Sterile inflammatory response. Cell-cycle. p53. Hyperproliferative stress response. Induced pluripotentianl cells (iPS).


RESUMEN

La regeneración hepática (RH) representa una de las formas más extraordinarias de respuesta al daño tisular. Debido a sus implicaciones terapéuticas, ha sido motivo de un interés extraordinario en las últimas décadas.
La RH es un proceso muy complejo y estrictamente regulado que cumple las características de los sistemas biológicos más evolucionados (robustness) y que explica la dificultad de interferir en ella con usos terapéuticos.
La RH reproduce el modelo fisiológico de respuesta al daño con una fase primera de preparación o "cebado" de los hepatocitos -transición G0-G1 del ciclo celular- y una fase ulterior de proliferación -fases S/M del ciclo- que termina con la recuperación de la masa hepática.
Dicho proceso se ha relacionado con los mediadores de la respuesta biológica al daño tisular: hormonas, sistema del complemento, plaquetas, citocinas proinflamatorias (TNF-α, IL-1β, IL-6), factores de crecimiento (HGF, EGF, VGF, etc.) y factores antiinflamatorios (Il-10, TGF-β).
Debido a su complejidad y a su estricta regulación, se explica que todavía hoy la única alternativa a la insuficiencia hepática sea el trasplante hepático.
La identificación reciente de las células pluripotenciales inducidas (iPS) y la capacidad plástica de las células madre mesenquimales (CD133+) ha suscitado nuevas expectativas a la terapia celular regenerativa. Dichos trabajos han confirmado la cooperación entre las células mesenquimales y epiteliales.
En el presente trabajo se describen los mecanismos fisiológicos de la regeneración hepática.

Palabras clave: Regeneración hepática. Respuesta inflamatoria estéril. Ciclo celular. p53. Respuesta estrés hiperproliferativo. Células pluripotenciales inducidas (iPS).


Abbreviations
AP-1: Activating protein-1.
Abs: Biliary acids.
ATP: Adenosine 5-tryphosphate.
AR: Amphiregulin.
ARNm: Messenger ribonucleic acid.
Cdc25b: Protein phosphatases that dephosphorylates Cdks.
Cdk: Cyclin-dependent kinase.
CD117: Hematopoyetic progenitor cells.
CD133: Endothelial progenitor.
Ckls: Cdk-cyclin complex inhibitor.
Cl4C: Carbon tetrachloride.
C-met: HGF receptor.
CT-1: Cardiotrophin-1.
C3a: Proteolitic fragment "a" from C3 complement protein.
C3b: Proteolitic fragment "b" from C3 complement protein.
C5a: Proteolitic fragment "a" from C5 complement protein.
Check-point R: Cell-cycle "restriction" check-point.
DAMP: Damage-associated molecular pattern.
DNA: Deoxyribonucleic acid.
EGF: Epidermal growth factor.
EGFR: Epidermal growth factor receptor.
EM: Extracellular matrix.
FGF: Fibroblast growth factors.
FXR: Farnesoid X receptor.
gp 130: Glycoprotein 130.
GSK3β: Glycogen synthase kinase 3b.
G0: Quiescent or resting cell state.
G1: GAP 1 phase. Period within cell-cycle following mitosis.
G2: GAP 2 phase. Period within cell-cycle between the end of DNA synthesis and the beginning of M phase.
HB-EGF: Heparin-bound epidermal growth factor.
HGF: Hepatocyte growth factor.
HIF-1α: Hypoxia induced transcription factor-1α.
HMGB1: High mobility group Box 1.
Hsp: Heat shock proteins.
IkbKβ: Cytoplasmic NF-Kβinhibitor.
IL: Interleukin.
IL-1β: Interleukin 1 β.
IL-6: Interleukin 6.
IL-18: Interleukin 18.
IL-6R/gp130: IL-6 glycoprotein 130 receptor.
iPS: Induced pluripotent stem cells.
JAK: Tyrosine kinase Janus or Janus kinase.
KO: Knout-out.
LPS: Lipopolysaccharide protein.
LR: Liver regeneration.
M phase: Cell-cycle mitosis phase.
MAPK: Mitogen-activated protein kinase.
Myc: Myc oncoprotein.
NF-Kβ: Nuclear factor Kappa β.
NK: Natural killer.
NKT: Natural killer T cell.
NLRP: NOD-like receptors.
NOD: Nucleotide digomerization domain - containing protein.
PAF: Platelet activator factor.
PAMP: Pathogen associated molecular patterns.
PDGF: Platelet derived growth factor.
PHx: Partial hepatectomy.
p53: p53 protein.
ROS: Oxygen free radicals.
RRP: Patron recognition receptors.
RTK: Receptor tyrosine kinases.
RTT: Toll-like receptors.
S phase: Cell-cycle DNA replication: synthesis phase.
SOCS: Suppression protein of cytokine signaling.
SOCS-3: Cytokine signaling suppression.
STAT3: Signal transducer and activator of transcription.
TFG: Transforming growth factor
TNF-α: Tumor necrosis factor-α.
TNF-R: Tumor necrosis factor receptor.
TPM1: Tryptofan hydrolasa.
VEGF: Endothelial-vascular growth factor.

 

"Nature adapts the organ to the function and not the function to the organ"
Aristotle, on The parts of the animals

 

Background

Liver regeneration (LR), is one of the most enigmatic and fascinating phenomenon of the animal scale. The rapid volume and function recovery following a major liver resection (> 70 %) or liver injury and its strict regulation at the initiation and termination periods is an exclusive characteristic of the liver (1-9).

In addition, LR is the scientific background of several clinical procedures as the extended liver resections (> 70 % of the liver parenchyma or five segments), split liver transplantation, liver donor procedure, two-stage hepatectomies, isolated portal vein embolization or associated with "in situ" liver transection, the artificial liver support in acute hepatic failure and the clinical application of cellular therapy as well (10-21) (Fig. 1).

 

 

Albeit the liver regenerative ability was described in the punishment inflicted to Prometheus by Zeus, the first scientific description was reported in 1879 (22-24). This ability represents one of the most extraordinary physiological response in maintaining the internal homeostasis. Besides the myriad of metabolic functions, the liver is the major immune organ and the first natural filter for micro-organisms (bacteria, virus) and xenobiotics originated in the intestines (25-28). In the last 50 years has been an extraordinary interest in knowing the underlying mechanisms of the LR, and the mentioned clinical applications (4,7,29-31).

In the present article we review the physiologic basis underlying LR and its relations with clinical conditions.

 

Liver regeneration characteristics

In basal conditions the hepatocytes remain in quiescent state (phase G0 of the cell cycle), but retain the ability to reinitiate the cell cycle with occasion of tissue damage, tissue loss or exogen stimulus (growth factors, mitogens) (32-35). LR has been studied in diverse experimental models; from cellular cultures to "in vivo" induced liver injury by toxics (Cl4C, galactosamine, thioacetamide), bacterial particles (LPS), virus and surgical models as partial hepatectomy (PHx) or segmental liver transplantation. Knock-out (KO) and knock-in animals with a genetic over-expression or a specific gen deletion have been used too (2,5,36-38). Others authors have investigated the gen expression profile with microarray techniques (39-43). Some of the mentioned models are summarized in table I.

 

 

From the above mentioned models highlight those in which the liver volume, function and survival were evaluated. Yet the KO models have been crucial for the signal pathway identification, their assessments have been difficult because LR pleiomorphic feature (40).

The 70 % PHx procedure in rodents and in big animals as well, is the most used, because represents the major regeneration stimulus and elicits the synchronic and homogeneous response of the remnant liver (36-38). This model has allowed to compare the regeneration between control animals (wild type) with the genetic modified on signalizing molecules, receptors and cell cycle regulators (2,5,44).

The liver mass recovery following a PHx or after an acute injury is achieved by a compensatory hyperplasia in contrast with a true epimorphic regeneration of the lost tissue; that takes place in the inferior vertebrate-Zebra fish, Salamander or amphibians which are able to renew the removed limbs or other anatomical structures-major appendages, inferior jaw, portions of intestine, cardiac ventricle along the life time (45-49). These animals regenerate the lost tissue from the "blastema" formed by the transdifferentiaton of adult cells into mesenchymal cells (45,47,48).

The liver regeneration characteristics can be summarized in the following:

1. It is a phenomenon present in all vertebrates from Zebra fish to the human; in whom common mechanisms are involved: Thrombin activation, innate immune system participation, ploidy and aneuploidy development, tissue remodeling and the strict regulation of liver volume at the end of the process known as "hepatostat" regulation (34,46,50,51).

2. LR represents a strict regulated response, according with the pattern depicted in figure 2. The hepatocyte duplication, the mesenchymal cells duplication sequences, the morphogenesis and volume reached at the end of the process are genetically regulated. It is phenomenon different form embryogenesis or wound healing although share common signaling and transcription pathways (4,49,52).

 

 

3. It is a very rapid and explosive process which initiates in seconds and ends in few days -depending on the age and species- with the recovering of the hepatic volume and involves the signaling mechanisms -autocrine, paracrine, endocrine- of cell proliferation, morphogenesis, liver zonation and metabolism regulation genes (1,2,4,5,37,44,53). In recent years, diverse signaling and transcription pathways have been described, many of them related with the physiologic liver response to cell injury. Herein we shall describe how LR process reproduces the tissue damage response pattern (54-56).

LR begins with the recognition of the pathogen-associated molecular patterns (PAMPs) and the damage-associated molecular patterns (DAMPs); triggering the natural immune response-C3a and C5a complement fraction activation, TNF-α, TNF-α secretion, IL-6, IL-1β, IL-18 synthesis-which induce the hepatocytes to proliferate. The transition from the resting G0 state to the G1 phase of the cell cycle (priming phase), comes about during the first 4 hours after liver resection (57-67).

4. The cell cycle phases are reproduced throughout the LR response, having arouse a great interest in knowing the gene expression of the regulatory mechanisms involved and its translation to other fields such as cancer (33,35,42). The cellular proliferation accounts in a chronological and sequential order; the hepatocytes divide first following by the kupffer, endothelial cells, vascular and biliary canaliculi neoformation just before the hepatic structure is replicated.

Early following a 70 % HPx; 95 % of the hepatocytes transient form the G0 phase to the G1 cell cycle phase (1,4,68); at the end of which exists the critical control point denominated "R" (restriction point), which decides if the cells divide irreversible. Until then, the process is reversible, allowing the hepatocytes to return to the previous state (G0) if the conditions were not favorable. Beyond this point the progression of the cell cycle is completed until the hepatocyte division is accomplished (69, 70) (Fig. 2).

An increasing in the preformed cytosolic preformed factors (TFs) -signal transducer and activator of transcription (STAT3), nuclear-Kappa b factor (NFKβ) and activating protein 1 (AP-1)- have been described in the first 30 min post-hepatectomy as consequence of the signaling transductions induced by cytokines bindings with their receptors. TFs trigger the protein synthesis and gene expression required for initiating the cell cycle (known as priming phase). DNA synthesis increasing (phase S) has been reported during the 24 and 36 hours post-hepatectomy in the rat and in mouse respectively. The hepatocytes enter into mitosis at 48 hours post-PHx, following by kupffer cells. The stellate and biliary epithelial cells enter into S phase 48 hours post-hepatectomy and proliferate in slow pattern. Endothelial cells proliferate in the third day showing a peak at the fifth post-PHx day (3,4,42,68,71-73).

Additionally, a hepatocyte population return to the resting state (G0) at 72 hours post-hepatectomy meanwhile other reinitiate the cell division until reaching the original liver size (4,6,72). It has been postulated, that the second proliferative wave is due to growth factors secreted by the own hepatocytes and to the additive effect of co-mitogens such as norepinephrine, insulin, somatostatin and glucagon (1,2,4).

Miyaoka et al. (52) has recently described that hepatocytes duplicate their size and increase the hepatocyte number by 1,5 fold, following PHx. These authors have reported an increase in the hepatocyte size by 1,6 and 0,7 divisions per cell suggesting that LR underlies two different processes: Hypertrophy and proliferation. The same authors observed in 30 % liver resection model the liver remnant increasing size was due to a significant 1,5-fold increase in the hepatocyte size without undergoing cellular division (52).

From the morphological point of view, several cluster of hepatocytes (10-14 mononuclear and binuclear ploidy cells -tetraploid (4n), octaploid (8n), 16n...) were observed shortly after PHx. A replication of the stellate and sinusoidal cells, increasing the size of liver lobules, was observed two-four post-hepatectomy days (35,74,75).

Ding et al. (76) has described the secretion of angiocrine factors and vascular neo-formation. The relative hypoxia of the residual liver-due to overactive hepatic arterial buffer response, stimulates the hypoxia-induced factor 1 alpha (HIF-1α) -within the first 12-48 post-hepatectomy hours- which activates the vascular-endothelial growth factor (VEGF), the fibroblast-growth factor (FGF) and the inducible nitric oxide (NO) synthase (77-81). Proliferation of hepatocytes proceeds from the periportal to pericentral areas of the liver lobule: Once the original size has been restored an apoptotic remodeling and zonalizing process takes place (42,44,72,82,83).

5. In contrast to other solid organs, as skin, or the intestines, the restoration of liver mass is achieved by the mature hepatocytes and mesenchymal cells proliferation neither by pluripotent stem cell differentiation or oval cells; although its participation has been described whenever hepatocyte division is impaired or abolished as occurs in chronic liver diseases (1,2,3,5,42,44,72,84-87).

With occasion of the oval cell-hepatocyte precursor description and the plasticity of the hematopoietic cells, endothelial progenitor cells (CD133, CD117) and the induced pluripotential cell (iPS) described by the 2012 Nobel Prize Shinya Yamanaka (88-90); an explosion of articles regarding cellular therapy in liver diseases have been published (91,92). Given the enormous number of papers, we refer to excellent reviews (93-97).

Despite the aroused expected outcomes, the results have shown limited efficacy so far. Even trials performed with isolated adult hepatocytes in inborn errors of metabolism the results are very limited. In the majority of cases only a temporarily improvement was achieved (20,98). Preliminary clinical trials using bone marrow growing factors (G-CSF) in acute-on-chronic liver failure, and bone marrow progenitor cells (CD133) in chronic disease have recently published (96-99). Clinical results as happened with other solid organs (heart, pancreas, intestine) suggest that tissue regeneration is indeed a complex process which depends of the cooperation between different cellular lineages (87,100-102).

6. The liver regenerative capacity is almost unlimited. Seven consecutive 50 % partial hepatectomies, without developing liver failure or preventing the regenerative response has been described in the rat (2-4). In a model of homozygote tyrosinemia (FAH -/-), the metabolic disorder was reverted after syngeneic hepatocyte transplantation. When the grafted hepatocytes were again isolated and transplanted they were able to reverse the enzymatic deficit in a FAH -/- second generation, and successively until 10 generations! It has been estimated that mouse hepatocytes could undergo up to 69 divisions, equivalent to generate 50 livers (103,104).

7. Another intriguing enigma of LR is the tight relationship between the hepatic volume and body surface; known as Liver Index (liver weight/body weight x 100 ~ 2.5 %); called as "hepatostat" regulation. In clinical and in experimental liver transplantation field, it is well known the adaptation of the donor liver size to the recipient body surface; small grafts in big recipients and vice versa. This phenomenon has even been reported in the baboon xenograft to the human (4,105-107).

Albeit most of the cellular changes throughout LR have been related with the cell nuclear changes; during cell division the organelles have to duplicate too. The regulation between cell growth and chromosome duplication is poorly understood (69,108-110). The majority of growth factors (epidermal growth factor -EGF-, hepatocyte growth factor -HGF-, insulin growth factor -IGF-1-) besides to their mitogenic effect, they increase the growth rate and promote cell survival by inhibiting apoptosis (5,69,108).

In inferior vertebrates and mammals is common to observe ploidy hepatocytes, tetraploid (4n) or octaploid (8n), mononucleated or binucleated (2x2n, 2x4n, 2x8n) as consequence of incomplete cytokinesis (Figs. 3 and 4). Ploidy degree differs across many species. In rats, 80 % of hepatocytes are polyploid and aneuploid, 60 % in mouse and 30 % in human being (75,111). Ploydia's first consequence is the hepatocyte increasing size (2,112,113). It has been pointed out that cellular size relies upon the DNA nuclear amount. Likely an increase in the chromosome number correlates with an increase in cell size (114-116).

 

 

Following a hepatectomy in rodents a 20 % to 5 % decreasing in binucleated hepatocytes and an increasing in tetraploid-4n-and octaploid-8n-, and even up to 16n hepatocytes have been described at 72 hrs post-HPx; showing that HPx represents and intense stimulus for DNA replication before cytokinesis (2,112,116,117). In those animals in which chromosome separation was inhibited, the recovery of liver mass following HPx took place by the hepatocyte polyploidy response, 18n, 32n, and higher degree (118,119).

This phenomenon coined as endorreduplication gives rise to chromosome replicating sequences without cellular division, generating cells with several genome copies, enhancing gene expression (120). Duncan et al. (35,74,111) has recently described the ability of hepatocytes to develop polyploidia and its reversion to aneuploidy state as an adapting response to xenobiotics or dietetic stimulus. It is remarkable that the liver function remained stable throughout the process, revealing the functional equivalence between 2n, 4n, and 8n hepatocytes (2,112). Kurinna et al. (117) has recently published the p53 control role in quiescent hepatocyte as well as during regeneration (7 days). The authors described that KO mouse for p53 (p53 -/-), show a higher ploidy degree than control mouse (wild type, p53 +/+), and that they initiate earlier the cellular division, develop an increasing nuclei and cytoplasm size, and a higher degree of ploidy contrasting with p53 +/+ (wild type) after 70 % hepatectomy. p53 lacking impairs ploidy reversion, confirming that regeneration in p53 -/- rodents likely is based in cell size augmentation as well. The mentioned authors found more errors in chromosomal integrity in p53 -/- mouse (multipolar spindle, lagging chromosomes) than in control animals (p53 +/+); yet the recovery of liver mass at 7 days post-hepatectomy was equivalent (52,117,121,122).

8. Several authors have pointed out that a more intense liver damage (extreme liver resections > 70 %, living donors transplantation with small grafts < 30 %, submassive hepatic necrosis), induces a stronger proliferative liver response (52,113-116). In liver donor transplantation setting using smaller grafts (≤ 30 % of standard liver volume) an accelerating in graft regeneration has been described (123,124). This pattern is similar to the classical physiological stress response or to tissue damage response as well (32,125-132).

9. Despite its amazing metabolic complexity, the liver keeps the homeostatic functions-albumin, coagulation factors synthesis, xenobiotic clearance, etc., throughout the regeneration process. A selective enhancing of proliferative genes and a temporarily silencing of carbohydrates metabolic genes were reported in the first 40 post-hepatectomy hours (42,72). Studies with light microscopy have revealed periportal changes of the classical lobule structure, 24 hours after 70 % hepatectomy (34,42,72).

Three peaks of hepatocyte DNA synthesis was reported by Wu et al. (82), initially in zone 1 and later in the mid-zonal "zone 2" of Rappaport's classical lobule description. Approximately 15 % of hepatocytes do not enter into cell division meanwhile 11 % of them divide by three fold. The underlying mechanism of this phenomenon remains elusive.

10. Besides brain, the liver is the organ with major capacity of reacting to broad internal perturbations and keeping its structure and function as well. This quality is also present in several biological systems just as: Physiologic homeostasis, tissue development, cell-cycle, ecological resilience, circadian rhythm, and even cancer development (133-135). This phenomenon was coined as "robustness" and has concerned great interest in evolutionary biology in the last decade. LR shares all the robustness common features: Pleomorphism, redundancy and biofeedback mechanisms.

This makes that LR's global understanding is extremely difficult and as consequence therapeutic interventions on LR is still lacking (2,4,10,44).

 

Phases of liver regeneration: priming, progression and termination

It is a merit of Nelson Fausto (1,2), who has integrated the cellular and molecular mechanisms of LR in a three-phase model. LR can be divided into three phases: An initial phase of induction or priming phase which corresponds to hepatocytes transition from quiescent state, or G0 to G1 phase of the cell cycle, which takes place in the first 4 h post-HPx. A second phase or progression phase corresponds with the transition from G1 to the completion of mitosis; and a third phase or "apoptotic" phase of tissue remodeling and finally return to the initial G0 phase. The above model has been assessed in different species and has the virtue of corresponding with the cell-cycle phases, from which some basic aspects are reviewed (32,69,136-138).

Cell-cycle is divided into four phases: G1, S, G2 and M according with figure 5. DNA synthesis takes place in S phase (from synthesis). The chromosome segregation occurs in the M phase or mitosis, which is divided into four stages: Prophase, metaphase, anaphase and telophase (the cellular division ends with the cytokinesis giving rise to two daughter cells, which can reinitiate the cell cycle or return to the previous G0 state). G1 and G2 phases also known as gap phases take place before DNA synthesis (S phase) and mitosis (M phase) respectively, adding extra time for cellular growing and surveillance of the transition to the following phases according with the extracellular and intracellular signals (69,137-139).

 

 

The cell-cycle has three control checkpoints: restriction or "R" check-point which defines the entry into the cycle in late G1 phase; and the two mitotic control check-points: The G2/M check-point which regulates mitosis entry and the metaphase-anaphase transition control check-point where the final mitosis events are initiated giving rise to the sister chromatides and mitosis cessation (69,70,136,140,142). The control "R" check-point regulates the rate of cell division and determines the cell cycle irreversibility, at whatever time cell progress beyond "R" point (69).

The majority of mitogens and growth factors- such as, platelet derived growth factor (PDEGF), EGF, HGF, and transforming β growth factors regulate the rate of cell division in the "R" check-point, when the cell is more sensitive to external factors (138-140).

 

Initial priming phase

Sterile inflammatory response

According with the mentioned model, LR represents a physiological response to liver damage (63,143). The tissue injury can be expressed as "cellular stress" or as one of the knowing forms of cellular death: Necrosis, apoptosis or necro-apoptosis (58,144-147). Necrosis induces the recognition of DAMPs by the pattern recognition receptors (PRRs) which triggers the innate immune system. PRR's receptors have been described in the cell membrane, endososomal membranes and in the mesenchymal and hepatocytes cytoplasm as well (44,54,55,61,62,148,149).

The regenerative response could restore the threatening situation or fail, depending of tissue injury severity. In the latter condition, liver is unable to restore the homeostasis, being associated with a high mortality and requiring in most of the cases a liver transplant as definitive solution (example: Fulminant hepatic failure, small-for-size syndrome, acute-on-chronic liver failure, or post-hepatectomy liver failure) (150-152). In the above mentioned situations a progressive cholestasis, coagulopathy, encephalopathy, sepsis and multiple organ failure takes place. Some authors have related it with an innate immune "paralysis"; others have suggested that an excessive mitogenic stimulus could provoke a "hyperproliferation stress response" which leads to cellular apoptosis and failing liver (153-155).

In 1994 Polly Matzinger described the DANGER theory by which the natural immune system, besides recognizing pathogen germs and their wall components also recognizes the endogen ligands released by cellular damage and necrosis, giving rise to the denominated "sterile inflammation response" (54,55,59,156-158).

Necrotic cells leak the intracellular content -DAMPs-out to the extracellular compartment where bind to the patron receptors PRRs. Among those receptors belong the complement system and Toll-Like receptors (TLRS) which are expressed in the membranes and in the cytosol of macrophages, sinusoidal endothelial, dendritic cells, natural killer (NK) and hepatocytes (159-163) (Fig. 6). DAMPS are also recognized by a subsets of stress and damage receptors, known as NLRP which belong to the NOD-like receptors (NOD). Once they are triggered, give raise the "inflammosoma" complex, releasing pro-inflammatory and mitogenic cytokines such as: interleukin-1β (IL-1β) and interleukin 18 (IL-18). The NLRP receptor role has raised great interest in recent years regarding the mechanism of inflammation and survival (164-166).

 

 

Since the DANGER theory description by Matzinger, new tissue injury related DAPMs have been described just as: high mobility Group Box 1 (HMGB1), heat shock proteins (Hsps, Hsp60, Hsp70), uric acid, genomic DNA, ATP, adenosine heparin sulfate, oligosaccharides, degradation fragments of hyaluronic acid, mitochondrial N formilated peptides, which have been denominated with the general term as "alarmines" (59,63,167-169).

It is indicative that recognizing proteins involved in the innate immune system are phylogenetic previous to the separation between vegetals and animals (thousand of million years ago) and to the acquisition of the adaptive immune system. On account of the seminal description of TLRs in Drosophila Melanogaster and in humans, as well as the DANGER theory, the paradigm of the innate immune system response and tissue regeneration has been reported (170-176).

The interaction between TLRs and their ligands triggers several signaling cascades which generate IL-6, TNF-β, IL-1β synthesis, and the activation of cytosolic transcription factors (NF-Kβ, STAT-3, AP-1) conveying the pro-proliferative stimulus to the nucleus (62). TLRs regulate the regeneration process in colon intestinal mucosa, lung, skin and post-hepatectomy regenerative response by eliciting pro-survival signals and apoptosis inhibition too (63,170,177-179).

LR has also related with the endogenous ligands present in portal blood: Bile acids (BAs), xenobiotic, LPS and components of extracellular matrix-fibronectin, heparan sulfate, fibrinogen, hialuronic acid oligosaccharides- released by tissue injury (surgical manipulation) (180-185).

The nonparenchymal cells -specially macrophages-synthesize pro-inflammatory cytokines just as: IL-1β, TNF-α, IL-6, gamma-interferon (IFN-Y), protaglandins and activating platelet derived factor with antiapoptotic and proliferative functions (63,167,186-188).

TNF-α and IL-6 cytokines initiate the "priming" phase of hepatocytes (transition G0-G1 of cell cycle). These cytokines signalize through the TNF receptors (TNF-R1, TNF-R2 and IL-6 (IL-R/gp130). They are tyrosine kinase receptors similar to growth factor receptors, which lead to enzymatic cascades as the mitogen-activated protein (MAP) kinase cascades. Janus Kinase -JAK- is part of this group, which upon activation phosphorylates preformed transcription factor STAT-3 (JAK/STAT signaling). STAT-3 protein binds to the DNA activating the immediately-early response genes expression (IEGs): c-fos, c-jun, c-myc (known as oncogenes) which promote the cell-cycle entry (Fig. 6). More than 180 of these genes have been described, which synthesize the required protein to leave the quiescent G0 state (1,4,5,42,44,72,118,188-191).

The priming phase initiates in the first 30 minutes longing for the first 4 hours post-hepatectomy. In addition to STAT-3 factor, others involved transcription factors are Kappa-β nuclear factor (NF-Kβ) and the activating protein 1 (AP-1) required for the "novo" synthesis of G0-G1 and G1/S regulation proteins. The STAT-3, NF-Kβ and AP-1 participation were assessed in conditional KO animals, with genetic manipulation techniques (192-195).

Animals with mutations in TNF-α receptor (TNF-R1), showed an inhibition of NF-Kβ transcription and a severe deficit in liver regeneration, confirming that NF-Kβ transduction is crucial in the Kupffer cells response to cytokines. KO mouse for IL-6 receptors (gp 130) showed minor defects in cellular proliferation. In KO animals for IL-6 (IL-6 -/-) and its receptor (gp130), LPS administration following a partial hepatectomy significantly decreased the survival; confirming that IL-6 has a protector role in regeneration. Hepato-specific deletion of STAT-3 and AP-1, decrease the cyclin expression; which is mandatory for the G0-G1 transition in the cell cycle. On the other hand, when NF-Kβ factor was up-raised by blocking its cytoplasmatic inhibitor IkbKβ, a more intense inflammatory and proliferative response was assessed (72,196).

In addition to the mentioned inflammatory mediators-TNF-α, IL-6, C3a, C3b, LPS, growth factors as HGF, PDGF and EGF, also stimulate G0-G1 transition. The HGF signalizes through the C-met receptor, and activates the immediately early response genes. Mouse with a conditional mutation in Met receptor, showed a defect in LR (6,197,198).

As it will be described in the proliferation phase, HGF regulates gene expression of other cell cycle phases and exerts "pro-survival" effects by inhibiting apoptosis (4).

Complement system and liver regeneration

In mouse models of liver injury Cl4C, partial hepatectomy and in patients submitted to a hepatectomy, a rising in the activated fractions of C3a and C5a were described in the first 24 hours post-procedure (131,132,149). Deficient mouse (KO) for C3 fraction (C3 -/-) and C5 fraction (C5 -/-), showed an impaired regeneration after 70 % hepatectomy. In these animals a decrease in the TNF-α, and IL-6 mRNA levels were assessed, in addition to a reduction in NF-Kβ and STAT-3 transcription factors (172-174). C5a receptor (C5aR) antagonist administration, provoked similar effects increasing the mortality; verifying the role of the complement in the initiation of the priming phase (199).

Both C3 -/- and C5 -/- KO animals developed more severe liver lesions than control animals following Cl4C infusion or following a PHx. The regeneration impairment and hepatocellular injury were restored when C3a and C5a were administered. The same phenomenon was observed in double knockout animals (C3/C5 -/-) as well as regeneration recovering after C3 and C5 fractions administration.

Lambris's group (170) has reported that C3a fraction triggers the IL-4 synthesis by NKT cells in the first 24 hours post-PHx and elicits the complement protein synthesis by macrophages during the priming phase. Besides the elimination of pathogens and tumor cells, complement system represents the more speed humoral component for recognizing tissue injury through DAMPs exposure and triggering the reparative response (170,199).

Platelets and liver regeneration

In addition to the well known haemostatic functions, platelets express Toll-like-receptors, 2, 4 and 9, and participate in the innate immune response. These receptors recognize the previous mentioned PAMPs and DAMPs, by which some authors refer them as the "circulating guardians" of tissue injury similar to the hemocyte vestige in the invertebrates (200,201).

Platelets contain fibrinogen, Von Willebrand factor, adhesion proteins, angiogenic factors, hepatoprotective factors (VEGF, PDGF, HGF, IGF, EGF-1 and TGF-β) and 95 % of the circulating serotonin! Although they are non-nucleated cells, they have ARN and retain the synthesis capacity of more than 300 different proteins, among highlights the TGF-β (202,203).

In 1996 Tomikawa et al. (204) described that the thrombocytosis induced by a splenectomy significantly enhanced liver regeneration in rodents. Same findings were confirmed by other authors as well the opposite effects secondary to thrombocytopenia. The same group described an improving survival in a model of sublethal hepatectomy (> 90 % resected parenchyma) model by inducing thrombocytosis. Other groups have related platelets with a protective effect on the post-hepatectomy liver dysfunction and operative mortality with a threshold platelet count < 100,000/uL. The mentioned authors related these outcomes with the above cited growth factors and the insulin-growth factor-1 (IGF-1) (205,206).

Lesurtel published in 2006 the enhancing effect of serotonin in hepatic regeneration "in vivo" although its mitogen effects were already known in fibroblasts and hepatocytes "in vitro" (207). The confirmation of these findings were reported by the same group using KO animals for serotonin synthesis (TPM1 -/-) as well as improving survival following the administration of synthetic serotonin agonist (5HT2B) in a small-for-size murine model. The authors reported an increase of the endothelial fenestrations of grafts in the treated animals (208).

The same authors described the sinusoidal "pseudocapillarization" reversion in old animals as an increase in the survival rate following the agonist administration after PHx; suggesting that opening of the sinusoidal-endothelial fenestration would facilitate the direct contact of platelets, cytokines and growth factors with the hepatocytes (208). Others studies have related the serotonin proliferative effect with VEGF synthesis. Although Matondo et al. (209) did not observe any effect in a murine model of deficient membrane carrier, KO (TOH1 -/-) after 70 % PHx.

Cytokines, growth factors and hormones

In 1967, Moolten and Bucher reported that the plasma from previously hepatectomized rats induced mitogenic effects on hepatocytes "in vitro" as well as "in vivo". Since then, several cytokines, growth factors, hormones and metabolites have been associated with the hepatocytes and mesenchymal cell cycle (4,210-213).

Family IL-6 cytokines

The IL-6 is an interleukin type I, associated with the innate immune response. Due to its cytoprotective, proliferative and anti-apoptototic effects is the most related with LR. In addition to IL-6 another interleukins sharing the Tyr-Kinase-coupled gp80 and gp130 receptors and signal transducing pathway have been described and referred as IL-6 family cytokines: IL-6, interleukin-11, leukemia-inhibitory factor (LIF), oncostatin (OSM), ciliary neurotrophic factor (CNTF), cardiotrophin-1 (CT-1) and IL-27 (186-188,214-217).

IL-6 is rapidly secreted by macrophages during the initial injury tissue response. IL-6 exerts local effects on hepatocytes as well systemic-fever, somnolence, ACTH and vasopressin secretion -inherent to the stress inflammatory response. IL-6 induces the acute-phase protein synthesis by the liver (reactive C-protein, A-amiloid) and exerts similar inflammatory effects to TNF-α and IL-1 (218-221).

IL-6 binds to gp130 membrane receptors, provoking the dimerization of two gp130 subunits (222). Each cytokine receptors associate with cytoplasmic Tyr-Kinases of the Janus Kinase family, termed Janus type (JAK1, JAK2, JAK3 and Tyk2-Tyrosine Kinase 2). Thereupon gp130 dimerization and its binding to the receptor (IL-Rα) promote JAK kinase activation which activates the STAT-3 (Signal Transducers and Activators of Transcription) transcription. The STAT proteins (7 in the human being) localized in the cytoplasm, migrate inside the nucleus, bind to the DNA promoter and activate the transcription genes related with cell proliferation, survival/apoptosis (223,224) (Fig. 6).

The activated STAT-3 induces the transcription of immediate-early genes c-jun, c-myc, c-fos and the early growth (EGR-1). Moreover the IL-6/KAK-STAT-3 signaling pathway provides the antiapoptotic protein Bcl-2, Bcl-x synthesis. The KAK-STAT pathway is one of the more straightforward strategy of signaling transduction from the cell membrane to the genome. Hepatospecific deletion of STAT-3 decreases the cyclin D1 and E1, both required for the cell cycle entry (72,191,192).

Besides STAT-3, there are two other transcription pathways mediated by Tyr-kinase receptors, the nuclear factor kβ (NF-Kβ) and the activating protein 1 (AP1). The NF-Kβ factor usually remains inactive in the cytoplasm by the inhibitory protein IkbKβ. The cytokine binding to the transmembrane receptor triggers IkbKβ degradation, releasing the NF-Kβ factor which translocates to the nucleus and stimulates the acute phase protein synthesis and the mentioned pro-proliferative and antiapoptototic genes. A rapid increase in TNF-α and IL-6 levels takes place after a 70 % PHx, or secondary to liver injury (ischemic, Cl4C, LPS administration). KO mice for IL-6 (IL-6 -/-) suffered hepatic failure following PHx, which was reversed upon previous administration of IL-6 (225). STAT-3 activation was almost absent in IL-6 -/- animals.

The STAT-3 protective effect on tissue injury was confirmed in KO conditional models, following adenovirus exposure (otherwise STAT-3 model is lethal in the embryo period) (198). The IL-6 cytoprotective effect was reported in diverse murine models of hepatotoxicity as ischemic-reperfusion injury or severe cholestatic. The IL-6 antiapoptotic effect was recently proved in extreme hepatectomies (87 % parenchyma resected) as well as in the rapid regeneration of "small-for-size" syndrome model (226-229). The AP-1 transcription factor promotes the immediately-early genes expression during the first five posthepatectomy hours; particularly the Jun oncoprotein which facilitates the G0-G1 transition of the cell cycle (72).

One of the IL-6 family cytokines, which has concerned particular interest is the cariotrophin-1 (CT-1). Prieto's group has described its protector effect in apoptosis as well as a regenerative inducer in ischemic-reperfusion injury, in an extreme PHx (92 % resection) and in fulminant hepatic failure experimental model (230-232).

Metabolic and xenobiotic signaling receptors

In addition to the extraordinary capability of the hepatocytes for entering into the cell cycle, meanwhile the specific metabolic functions ought to be performed by the remnant liver along the process (2,4,5,42,72). The related metabolic gene expressions are initially supplanted by the genes involved in the cytoskeletum formation, the mitotic spindle assembly and mitosis. Transcriptional studies have shown a temporary silencing of the metabolic genes (first 40 post-hepatectomy hours) and a posterior recovering towards the end of mitosis (42,233).

It has been previously described that the bile acids, drugs detoxification and carbohydrates metabolism take part in the early priming phase. The tightly regulation of bile acids pool by the liver through the enterohepatic circulation is very well known (234). This pool is maintained in the human between 2 and 4 g, circulating about 12 times per day; in the mouse is kept in 4 mg (235). The bile acid increasing damages the cell membranes and mitochondrial too, promoting cell apoptosis and necrosis (236,237).

The bile acids homeostasis, is regulated by nuclear receptors, through "Farnesoid X-receptor" (FXR). Free and conjugated bile acids binding with FXR, stimulates the transcription factors involved in early regeneration process (G1-S transition) as FoxM1b and proliferative genes (Cdc 25) (166,180).

At the time of a 70 % PHx an abrupt increase in the portal flow to the remnant liver (1/3 of original mass) undergoes increasing by three fold the nutrients, bile acids and all contents present in the splachnic circulation (2,3,4,180,238-240).

Deficient mice in FXR suffer a delay in LR and higher mortality rate following PHx (180). These animals are unable to activate the early response genes c-myc, c-fos and c-jun. On the other hand the bile acids administration stimulated LR in previously hepatectomized mice and was delayed after cholesteramine administration.

The bile acids stimulate the pro-inflammatory cytokines as TNF-α and IL-1 by the kupffer cells with pro-survival, anti-apoptotic and proliferative effects (240). Bile acids and xenobiotics could behave as true DANGER molecules triggering the priming phase in addition to their mitogen effect, particularly the hydrophobic.

There is evidence that the administration of FXR receptor agonist enhance LR in old animals. These findings as their cytoprotective properties have raised its likely clinical application (241,242).

The relationship between the metabolic pathways and regeneration is complex given that the metabolic variations could be a consequence of the silencing of metabolic genes process without implying a direct mitogen effect. By the way, some authors have reported that persistent hypoglycemia in hepatectomized mice stimulates regeneration, meanwhile the glucose addition inhibits LR (42,243). Those findings seem to be contradictory with Fissette (244,245), who reported that glucose and insulin infusion previous to a major resection improved the liver dysfunction and regeneration.

Liver ability to response to endogen stimuli -bile acids- and xenobiotics is an exclusively feature of the liver which could explain its almost unlimited capability for a "robust" response to a wide range of stimuli as liver resection, ischemia or bile acid overload (4,42,240).

 

Progression phase

Mitogenic signal transduction

Although there is not a clear limit between the LR phases; the progression phase takes place since the end of the G1 phase to the cellular division or cytokinesis (2,4,5,9). Besides the chromatine, other cytoplasm organelles -mitochondria, Golgi apparatus, lysosome, endoplasmic reticulum- ought to duplicate during this period in order to maintain the ratio between the cytoplasm and nucleus size in advance of accomplishing the metabolic functions of the liver (69,73,109,110).

The progression phase is regulated by the previous mentioned growth factors -HGF, EGF, VEGF, and TGF-a- which as well their mitogen effects, exert trophic and pro-survival functions.

The primary growth factors role is to activate the Cdks-cyclin complex which evokes a wide variety of cell cycle changes: Mitotic spindle duplication and DNA replication in the S phase. The majority of the growth factors convey the mitogen signal to the nucleus through the membrane and cytoplasmic tyrosine-kinase receptors (RTK) promoting the downstream kinase cascade. The MAP-kinase cascade induces the immediately-early genes expression (c-fos, c-jun, c-myc), which is the first transcriptional event (two hours post-PHx). Among the factors codified by the pro-mitotic genes highlight FosM1b and Myc factors (1-6,42,43,246).

The FosM1b factor increases in the first post-PHx hour and evokes the activation of a large family of transcription factors termed AP-1, which stimulates the late response genes expression (two-days post-PHx). These genes regulate G1 cyclins (D1, D2, D3) required for the entry in the cell cycle (42). KO mice for c-jun, show 50 % mortality after PHx. The regeneration impairment is associated with an increase in cell necrosis and hepatocyte lipid accumulation. An increasing in the inhibitory G1-S transition protein has been described.

The Fos M1b factor promotes the late response genes expression (2nd post-PHx day) and the G2-M transition. The role of this factor in the entry into mitosis and in the chromosome segregation was confirmed in hepato-specific FosM1b KO mice (247). It has been reported that hepatocyte over-expression of FosM1b restored the injured liver, more efficiently than control hepatocytes (wild type), suggesting eventual therapeutic applications in old patients with liver diseases (248).

The Myc factor remains elevated throughout the entire cycle and promotes the "R" control point transition as well as the gene expression involved in the cellular size and metabolism (108). The Cyclin D levels remains elevated throughout the cycle meanwhile the mitogen is present with independence of the cell cycle phase (108).

Growth factors and hormones such as insulin signalize through the Ras kinase cascade -MAP-, which phosphorylates and inhibits the glycogen synthase kinase (GSK3β) activity, allowing glycogen synthesis and also the activation of the preformed factors -AP-1, Myc- which stimulate cellular proliferation.

Hassanain et al. (244,245) has recently described the benefit effect of insulin and dextrose infusion in patients submitted to a major hepatectomy. These authors have assessed glycogen liver increasing as well as an improvement in the early postoperative liver function. Besides the glycogen protector effect as energy source for cellular division, GSK3β inhibition enhances the gene expression required for the regenerative response. The trophic effect of pancreatic hormones on liver parenchyma and liver regeneration was described by Starzl et al. in 1967 (249-252).

Hepatocyte growth factor (HGF)

The HGF was discovered in 1984 as a plasma protein with mitogenic effect in hepatocytes "in vitro", from hepatectomized rats and initially denominated "hepatopoietin" (235). Given its mitogen effects "in vivo" and "in vitro" is the growth factor, which has raised more attention. In addition to the mitogenic effect have been reported diverse effects as: Mitogenic (enhancement of cell motility), trophic, antiapoptotic, angiogenic and morphogenic effect on liver, brain, placenta, lungs, intestines, myocardium and reproductive system (211,253,254).

The HGF is synthesized by mesenchymal cells as a precursor (pro-HGF) molecule and stored in the extracellular matrix (ECM). HGF is a glycoprotein very similar to the blood coagulation cascade factors and fibrinolysis as well (plasminogen). Pro-HGF activation involves urokinase-type plasminogen activator (U-PA) (2,4,69,255-261).

Following a hepatic injury the plasma levels of HGF increase very rapidly by 10 to 20-fold. The HGF plasma raising is derived from the stored HGF and the releasing by the macrophage synthesis upon IL-6 and TNF-α stimulation. In addition HGF transcription increases in the mesenchymal cells of lung, kidney and spleen confirming its endocrine function (2,4,259,260,262-265). HGF binds to the tyrosine kinase receptor -C-met- which its phosphorylation is assessed during the first 1 to 15 minutes post-PHx, with the largest increase at 60 minutes. Following hepatic resection prolonged increasing in HGF plasma levels were observed, and remained elevated up to two weeks after liver resection. The higher HGF increasing has been reported in fulminant hepatic failure; this finding has questioned its therapeutic role in this condition. This paradoxical phenomenon -inability to stimulate cell proliferation with very high levels-, has been attributed to the C-met receptor inhibition by competitive effect by others ligands such as: IL-6, TGF-β1 or to the mentioned "hyperproliferative stress response" by which an excessive mitogen stimulus provokes an apoptotic response by p53 activation (61,266-270).

The HGF systemic and portal infusion in rodents increases DNA synthesis in the hepatocytes of zone 1. The infusion of human HGF into the portal vein in rodents resulted in hepatocyte proliferation and increase in relative liver mass (2,4).

The human HGF gene transfection by the hydrodynamic injection of plasmid DNA induces hepatomegaly in mouse via b-catenine activation, and the infusion of large amount of HGF increases the liver weight/body weight ratio associated with the mitogenic stimuli. The HGF withdrawal promoted apoptosis and the restoration of the basal DNA level (2,4,255,271).

The collagenase pretreatment in rats resulted in a higher response to HGF. Furthermore, isolated hepatocytes obtained by collagenase tissue digestion showed cell cycle initiating markers. This "pre-proliferative" pattern is according with the competence acquisition phenomenon described by Pardee in 1989; by which the competent hepatocytes response steadfast to a posterior regenerative stimulus (1,4,73).

It has been proposed that the releasing of remodeling metalloproteinases from the extracellular matrix would contribute to the initial regeneration phase, sensitizing hepatocytes to the HGF stored in the liver. Likely the extracellular matrix remodeling by the urokinase and metalloproteinases, would generate a sterile inflammatory response by releasing DAMPs from tissue injury and consequently the binding to Toll-like receptors and activation of the transcription factors -NF-Kβ, AP-1 and STAT-3- involved in cell cycle entry (54,55,59).

The C-met membrane Tyrosine-Kinase receptor is present in epithelial and in mesenchymal cells. The C-met receptor binding with its ligand leads to down-stream kinase cascade activation by mitogens (MAP kinase); which stimulates the cytosolic transcription factors (AP-1) related with proliferation and cell survival.

In addition HGF also activates the Janus Kinase (JAK) pathway which activates the transcription factors such as STAT-3, nuclear factor Kb (NF-Kβ) and β-catenin. The HGF binding with C-met receptor phosphorylates b-catenin giving rise to its nuclear translocation and up-regulation of target genes such as cyclin D required for the G0-G1 transition of cell cycle (2,4,188,264,267,268).

The role of HGF and its receptor in the regenerative response was also investigated using transgenic models. Mice lacking HGF and C-met receptor (null mice) die during gestation and express a reduced live size and extensive loss of parenchymal cells (270). Two independent groups have investigated liver regeneration using conditional C-met KO mice. In these animals a reduction in DNA synthesis associated with an absent of kinase mitogen activated (MAP) signaling was observed following a partial hepatectomy. Also Cl4C administration impaired regeneration and the inflammatory changes were more severe in these animals (197,271).

Epidermal growth factor (EGF)

The EGF represents a family factors, from which highlight: Epidermal growth factor (EGF), amphiregulin (AR), heparin-binding EGF (HB-EGF) and transforming growth factor-α (TGF-α). These factors are over-expressed during a liver injury. They signalize through the tyrosine kinase receptors (RTK): EFFR/ErbB1, HER2/ErbB2, HER3/ErbB3 and HER/ErbB4, that are collectively called EbbB1-4, which upon ligand binding promote downstream the mitogen activated cascade (MAPK kinase) (4,73,272).

Mouse lacking EGFR die between midgestation and postnatal day 20 with severe defects in the placenta, brain, skin and lung (273). In animals in which EGF was blocked and a partial hepatectomy was carried out; 1/3 died and the survivors showed a delay in hepatocyte division (274). However the liver regeneration was accomplished, suggesting that EGF signaling is important but not crucial for LR (61,274).

The rest of the EGF family are also synthesized as pro-factors that remain attached to the hepatocyte membrane and released by proteolysis. After PHx, the plasma levels of EGF rise, resulting in an increased EGF/EGFR ratio, confirming that EGFR plays a mitogenic role in the initial phase of LR (4).

The HB-EGF role was investigated in KO models and in transgenic animals as well. A delay in hepatocyte proliferation was observed in the first case and by contrast liver regeneration was stimulated in the second one. Animals lacking amphiregulin showed an impaired regenerative response, following a PHx (275,276).

The TGF-a is produced by the own hepatocytes -autocrine effect- and furthermore exerts paracrine effects on the endothelial and biliary cells. After HPx the TGF-α levels increases in the first 24-48 hours. The TGF-α addition to hepatocytes "in culture" and "in vivo" induces DNA synthesis and stimulates the transition through the control "R" point of cell cycle (279-281).

 

Termination phase and liver size regulation

Liver regeneration ends when the initial liver mass is restored and enabling it to carry out the liver functions (liver index ~ 2.5 %). The strict liver index regulation is one of the most intriguing feature of LR, because underlies a strict control at the end of the cell cycle and in the "neoformed" tissue remodeling process too (2,4,5,42,61,72,73).

The regeneration termination phase has been related with anti-inflammatory and pro-apoptotic cytokines as well as "hepatostat" factors such as IL-10, signaling cytokine suppressor proteins (SOCS-3), plasminogen activator inhibitor PAI-I and particularly the TGF-β (5,6,61,282).

Elegant studies using microarrays techniques in mouse and also in pigs have provided that the initial pro-mitotic gen over-expression is later supplanted by metabolic genes activation and afterwards by the cessation of proliferation and the returning to the previous quiescent state (34,41,61,72,73).

The transforming growth factor b family (TGF-β 1-3) comprises a number of cytokines related with cell differentiation and healing. They signalize through two receptors (type I and type II) which activate transcription regulators known as SMAD proteins (Small Mothers Against Decapentaplegic) which are translocated to the nucleus. In most of the tissues TGF-β inhibits proliferation at the G1 cell cycle phase (283-285).

Following a PHx an early increase in TGF-β ARNm has been described though a TGF-β transitory "resistance" has been reported as consequence of a diminishing in the hepatocyte receptors as well as inhibitors over-expression. This temporarily resistance resolves after DNA synthesis, restoring the TGF-β capability for inhibiting the cellular proliferation, and promoting the regeneration cessation (5-7 post-PHx days in the rat, three weeks in pigs) (286-288).

In transgenic animals and in those in which TGF-β was infused the anti-proliferative effects on different regeneration phases have been assessed, though no major changes were observed due to its pleomorphic feature. A delay in cellular proliferation was shown in transgenic mouse overexpressing TGF-β receptors (61,289,290). Significantly the knock-out mice for TGF-β receptor II (TGFBR2 -/-) showed and earlier and increased DNA synthesis in the first 120 h post-PHx, however no difference in the regeneration cessation was found with respect to control animals (wild type), suggesting that another DNA inhibitory pathways could be involved (291).

Other members of the TGF-β family are the activins, from which activin A is the most common. It was isolated in 1993 as a pro-apoptotic factor of hepatocytes "in vitro". The activin A promotes the intracellular Smad activation, like TGF-β, having been suggested that both share complementary functions. The injection of follistatin -an activin receptor antagonist- following PHx, induced an increasing in the liver weight and in the hepatocyte proliferation, supporting that activin A inhibits the proliferation and takes part in regulating liver regeneration termination (5,61,73,292).

In hepatocyte culture and in "bioartificial" models, the extracellular matrix plays a significant role in hepatocyte differentiation and proliferation (60,61,73). In spite of representing only 0.5 % of liver weight, the ECM plays an active role in the tissue injury response and regeneration. Besides storing growth factors -PDGF, TGF-β, VEGF, HGF- the ECM contains cytokines, metalloproteinases (collagenase, gellatinase), and macromolecules as fibronectine, laminine and collagen (type I, II, and VI) very reactive whenever the endothelial-sinusoidal barrier is damaged (184,185,293-298). After liver damage -surgical trauma, ischemia, toxic- the type urokinase plaminogen activator (μ-PA) and metalloproteinase are released, stimulating the growth factors liberation as well as the macromolecules degradation (fibrinogen, fibronectin, laminin) and eventually the DAMPs expression (DANGER theory) and promoting the sterile inflammation response and the hepatocyte priming phase (60,156,157,185).

The initial post-PHx proliferation associated with ECM degradation has been reported; giving rise to assembling nests of hepatocytes with deficient access to endocrine and paracrine factors, which in basal conditions inhibit cell division. The hypertrophic compiled hepatocytes require a posterior remodeling process up to get the initial hepatic index (72).

Michalopoulos group has corroborated the inhibitory effect of an integrin linked kinase (ILK) upon hepatocyte proliferation "in vitro" (ILK). Hepatoespecific KO animals for this integrin acquire hepatomegaly and enhanced hepatocyte proliferation (42,299). The hepatoespecific ablation of ILK impairs the regeneration termination. When these animals are submitted to a PHx the deficient livers in ILK reach a 58 % higher weight than the initial at 14 days post-HPx. The HGF and its C-met receptor expression increasing and the cell cycle inhibitor p27 diminution were observed (299-302).

Given the great interest in developing "bio-artificial livers" through the re-populating of previous decellularized matrix scaffold with hepatocytes and pluripotential cells, its development exploitation has been stimulated keeping in mind the increasing demand of other solid organs than the liver for transplantation as kidney, lung and heart (293-298,303,304).

p53 and regeneration control

Multicellular organisms contain cell cycle control mechanisms which react to stress situations -lack of nutrients, hypoxia, DNA damage- arresting the cell cycle or inducing cellular apoptosis (305,306).

The most prevalent system is the gen regulatory protein p53, also known as the "genome gatekeeper" by which cell responses to DNA damage or another stress conditions (307-311). Even though p53 is known as a tumor suppressor factor (more than 25,000 mutations have been reported in human tumors), p53 has recently raised great interest as regulator of LR ending (312).

Kurinna et al. has described that the gen p53 is necessary for the high grade ploidy and aneuploidy reverting, reported in basal conditions likewise during LR. Mouse with p53 lacking (p53 -/-) were associated with a higher ploydia grade (8n and 16 n) as a result of failures in the cell cycle final phase or cytokinesis. The same authors described for the first time that p53 regulates the expression of genes involved in the three cell division phases: Initiation-progression, division and restoration to the quiescent G0 state after PHx (117).

It is well known that p53 activation arrests cell cycle inducing apoptosis at the time of cellular stress conditions as severe hypoxia, acidosis or an excessive mitogenic stimulus. The latter is termed as "hyperproliferation stress response" (307,308) (Fig. 7), p53 promotes the expression of the cell cycle inhibitor p21 and the pro-apoptotic proteins (caspases), leading to a permanent cell cycle arrest and even cell death (307).

 

 

The Myc and Ras -oncoproteins- proliferative activity is only attainable whenever p53 is absent. The hyperproliferation stress response is the "in vivo" counterpart of the replicative senescence process described "in vitro". Culture cells after several divisions undergo a stable cell-cycle arrest induced by p53 rising. Cellular replicative senescence has been related with hyperproliferation stress -overactive Myc and Ras- or with non-physiological conditions given "in vitro": Deficient extracellular matrix components and inadequate levels of oxygen. Additionally cells lacking p53 proliferate endlessly and are therefore called "immortal" (313-317).

Although the hyperproliferative stress response has been related with protective mechanisms against cancer development, this physiologic response could be the underlying setting of clinical situations in which an excessive mitogenic stimuli takes place as "small-for-size syndrome", fulminant hepatic failure, post-hepatectomy liver failure or acute-on-chronic failure. In these conditions a large mitogenic stimulus is provoked by the endothelial damage, necrosis, parenchymal hemorrhage and hypoperfusion as a result of arterial vasospasm (150,151,318-320).

Today is still unknown why the liver regenerates in some cases and fails in others, developing an irreversible hepatic failure; not yet there is a clear limit regarding the minimal size remnant in liver resection or in liver-donor procedure (10,151,321-324). In spite of the myriad of papers regarding liver regeneration, its regulatory mechanism still remains as an enigma (2,4-6,9).

 

Conclusions and future perspectives

Liver regeneration still endures as one of the most amazing an enigmatic paradox of adaptation and response to keep the homeostasis. In the last decades has extensively investigated given its therapeutic consequences. LR is an extremely complex process, strictly regulated, which reproduces the tissue injury restoration, with an initial or priming phase -G0-G1 progress of hepatocyte cell cycle- and a posterior phase of proliferation-phases S and M- that ends with the hepatic mass restoration. This phenomenon has been related with hormones (insulin, glucagon), cytokines (TNF-α, IL-1β, IL-10), growth factors (HGF, EGF, VEGF) and in the last decade with the mesenchymal hematopoietic stem cells (CD 133+, CD117+) and in addition with oval cells.

The current description of developing structures with "hepatoid" phenotype derived from induced pluripotential stem cells (iPS) in rodents, has raised great concern as a future alternative to liver transplantation; although because the liver complexity should be seen with caution (325,326).

LR rigorously accomplishes the characteristics of the most regulated biological systems (robustness) -pleiomorfism, redundancy and feedback mechanisms- such as the cell cycle, natural immune response or circadian rhythm, which illustrates the difficulty in its guidance for therapeutic goals.

Because the liver functions complexity, the intents of definitive or temporarily supplying methods have been elusive, with exception of liver transplantation. Perhaps cell therapy development and acknowledge of liver regeneration molecular and physiological basis could get near the expected dream of using the own liver regenerative capacity in the treatment of liver disease, which at present only liver transplantation is a real option.

Santiago Ramon y Cajal aphorism, "It is fair to say that, in general, no problems have been exhausted; instead, men have been exhausted by the problems" is very opportune regarding liver regeneration unveiling efforts.

 

Acknowledgements

We gratefully acknowledge to Lydia Munarriz for manuscript editing and transcription. We thank to Fabiola de Goñi and Beatriz Urbelz for their contribution in researching data. The authors acknowledge to investigators from the Hepatology Division of CIMA (Center for Medical and Applied Research). The authors apologize to the many authors whose works have not been cited owing to space limitations.

 

References

1. Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology 2006;43:S45-53.         [ Links ]

2. Fausto N, Campbell J S. Liver regeneration. En: Arias IM, Alter HJ, Boyer JL, Cohen DE, Fausto N, Shafritz DA, et al, editores. The liver: biology and pathobiology. 5th ed. Oxford: Wiley & Sons; 2009. p. 549-65.         [ Links ]

3. Michalopoulos GK. Liver regeneration: alternative epithelial pathways. Int J Biochem Cell Biol 2011;43:173-9.         [ Links ]

4. Michalopoulos GK. Principles of liver regeneration and growth homeostasis. Compr Physiol 2013;3:485-513.         [ Links ]

5. Nevzorova YA, Trautwein C. Liver regeneration. En: Boyer TD, Wright TL, Manns MP, Zakim D, editores. Zakim and Boyer's hepatology: A textbook of liver disease. 6th ed. Philadelphia: Saunders Elsevier; 2012. p. 20-35.         [ Links ]

6. Taub R. Liver regeneration: from myth to mechanism. Nat Rev Mol Cell Biol 2004;5:836-47.         [ Links ]

7. Karp SJ. Clinical implications of advances in the basic science of liver repair and regeneration. Am J Transplant 2009;9:1973-80.         [ Links ]

8. Curado S, Stainier DY. deLiver'in regeneration: Injury response and development. Semin Liver Dis 2010;30:288-95.         [ Links ]

9. De Jonge J, Olthoff K M. Liver regeneration: Mechanisms and clinical relevance. In: Jarnagin WR, Belghiti J, Büchler MW, Chapman WC, D'Angelica MI, DeMatteo RP, et al, editors. Blumgart's surgery of the liver, biliary tract, and pancreas. 5th ed. Philadelphia: Elsevier; 2012. p. 87-101.         [ Links ]

10. Clavien PA, Petrowsky H, DeOliveira ML, Graf R. Strategies for safer liver surgery and partial liver transplantation. N Engl J Med 2007;356:1545-59.         [ Links ]

11. Adam R, Laurent A, Azoulay D, Castaing D, Bismuth H. Two-stage hepatectomy: A planned strategy to treat irresectable liver tumors. Ann Surg 2000;232:777-85.         [ Links ]

12. Abdalla EK. Portal vein embolization (prior to major hepatectomy) effects on regeneration, resectability, and outcome. J Surg Oncol 2010;102:960-7.         [ Links ]

13. Dutkowski P, De Rougemont O, Mullhaupt B, Clavien PA. Current and future trends in liver transplantation in Europe. Gastroenterology 2010;138:802,9.e1-4.         [ Links ]

14. Brown RS, Jr. Live donors in liver transplantation. Gastroenterology 2008;134:1802-13.         [ Links ]

15. Vagefi PA, Parekh J, Ascher NL, Roberts JP, Freise CE. Outcomes with split liver transplantation in 106 recipients: The University of California, San Francisco, experience from 1993 to 2010. Arch Surg 2011;146:1052-9.         [ Links ]

16. Schnitzbauer AA, Lang SA, Goessmann H, Nadalin S, Baumgart J, Farkas SA, et al. Right portal vein ligation combined with in situ splitting induces rapid left lateral liver lobe hypertrophy enabling 2-staged extended right hepatic resection in small-for-size settings. Ann Surg 2012;255:405-14.         [ Links ]

17. Lafuente S, Bertran MJ, Escorsell A. Artificial liver support. Literature review. Med Clin (Barc) 2011;136:484-7.         [ Links ]

18. Gupta S. Stem cells and hepatocyte transplantation. En: Boyer TD, Wright TL, Manns MP, Zakim D, editors. Zakim and Boyer's hepatology: a textbook of liver disease. 6th ed. Philadelphia: Saunders Elsevier; 2012. p. 157-70.         [ Links ]

19. Thorgeirsson SS, Grisham JW. Hematopoietic cells as hepatocyte stem cells: A critical review of the evidence. Hepatology 2006;43:2-8.         [ Links ]

20. Dhawan A, Puppi J, Hughes RD, Mitry RR. Human hepatocyte transplantation: Current experience and future challenges. Nat Rev Gastroenterol Hepatol 2010;7:288-98.         [ Links ]

21. Shafritz DA, Oertel M, Dabeva M D, Grompe M. Liver repopulation by cell transplantation and the role of stem cells. En: Arias IM, Alter HJ, Boyer JL, Cohen DE, Fausto N, Shafritz DA, et al, editors. The liver: biology and pathobiology. 5th ed. Oxford: Wiley & Sons; 2009. p. 577-95.         [ Links ]

22. Esquilo. Prometeo encadenado. En: Esquilo, Sófocles, Eurípides, editores. Obras completas. Madrid: Cátedra; 2004. p. 85-126.         [ Links ]

23. Tiniakos DG, Kandilis A, Geller SA. Tityus: A forgotten myth of liver regeneration. J Hepatol 2010;53:357-61.         [ Links ]

24. Tillmanns H. Experimentelle und anatomische Untersuchungen über Wunden der Leber und Niere. Ein Beitrag zur Lehre von der antiseptischen Wundheilung. Arch Pathol Anat Physiol Klin Med 1879;78:437-74.         [ Links ]

25. Knolle P. Liver and immune system. En: Boyer TD, Wright TL, Manns MP, Zakim D, editors. Zakim and Boyer's hepatology: A textbook of liver disease. 6th ed. Philadelphia: Saunders Elsevier; 2012. p. 129-41.         [ Links ]

26. Crispe IN. The liver as a lymphoid organ. Annu Rev Immunol 2009;27:147-63.         [ Links ]

27. Gillilland MG, Young V B, Huffnagle G B. Gastrointestinal microbial ecology with perspectives on health and disease. En: Johnson LR, Ghishan FK, Kaunitz JD, Merchant JL, Said HM, Wood JD, editors. Physiology of the gastrointestinal tract. Vol.1. 5th ed. Amsterdam: Elsevier; 2012. p. 1119-34.         [ Links ]

28. Gonzalez-Navajas JM, Frances R, Such J. Bacterial DNA in patients with cirrhosis and sterile ascites. Its role as a marker of bacterial translocation and prognostic tool. Rev Esp Enferm Dig 2007;99:599-603.         [ Links ]

29. Jia C. Advances in the regulation of liver regeneration. Expert Rev Gastroenterol Hepatol 2011;5:105-21.         [ Links ]

30. Riehle KJ, Dan YY, Campbell JS, Fausto N. New concepts in liver regeneration. J Gastroenterol Hepatol 2011;26(Supl. 1):203-12.         [ Links ]

31. Pahlavan PS, Feldmann RE,Jr, Zavos C, Kountouras J. Prometheus' challenge: Molecular, cellular and systemic aspects of liver regeneration. J Surg Res 2006;134:238-51.         [ Links ]

32. Morgan DO. The cell cycle. En: Morgan DO, editor. The cell cycle: Principles of control. London: New Science Press; 2007. p. 1-9.         [ Links ]

33. Satyanarayana A, Geffers R, Manns MP, Buer J, Rudolph KL. Gene expression profile at the G1/S transition of liver regeneration after partial hepatectomy in mice. Cell Cycle 2004;3:1405-17.         [ Links ]

34. Chauhan A, Lorenzen S, Herzel H, Bernard S. Regulation of mammalian cell cycle progression in the regenerating liver. J Theor Biol 2011;283:103-12.         [ Links ]

35. Duncan AW. Aneuploidy, polyploidy and ploidy reversal in the liver. Semin Cell Dev Biol 2013;24:347-56.         [ Links ]

36. Higgins GM, Anderson RM. Experimental pathology of the liver. 1. Restoration of the liver of the white rat following partial surgical removal, Arch Pathol 1931;12:186-8.         [ Links ]

37. Mitchell C, Willenbring H. A reproducible and well-tolerated method for 2/3 partial hepatectomy in mice. Nat Protoc 2008;3:1167-70.         [ Links ]

38. Martins PN, Theruvath TP, Neuhaus P. Rodent models of partial hepatectomies. Liver Int 2008;28:3-11.         [ Links ]

39. Fausto N. Lessons from genetically engineered animal models. V. Knocking out genes to study liver regeneration: present and future. Am J Physiol 1999;277:G917-21.         [ Links ]

40. Mitchell C, Gilgenkrantz H. Transcriptional profiling of liver regeneration: New approaches to an old trick! J Hepatol 2003;38:847-9.         [ Links ]

41. Fukuhara Y, Hirasawa A, Li XK, Kawasaki M, Fujino M, Funeshima N, et al. Gene expression profile in the regenerating rat liver after partial hepatectomy. J Hepatol 2003;38:784-92.         [ Links ]

42. Kurinna S, Barton MC. Hierarchies of transcriptional regulation during liver regeneration. Prog Mol Biol Transl Sci 2010;97:201-27.         [ Links ]

43. Locker J, Tian J, Carver R, Concas D, Cossu C, Ledda-Columbano GM, et al. A common set of immediate-early response genes in liver regeneration and hyperplasia. Hepatology 2003;38:314-25.         [ Links ]

44. Fausto N, Campbell JS, Riehle KJ. Liver regeneration. J Hepatol 2012;57:692-4.         [ Links ]

45. Call MK, Tsonis PA. Vertebrate limb regeneration. Adv Biochem Eng Biotechnol 2005;93:67-81.         [ Links ]

46. Matthews RP. Zebrafish as a model system for the study of liver development and disease. En: Arias IM, Alter HJ, Boyer JL, Cohen DE, Fausto, Shafritz DA, et al, editors. The liver: biology and pathobiology. 5th ed. Oxford: Wiley & Sons; 2009. p. 1067-74.         [ Links ]

47. Carlson BM. Some principles of regeneration in mammalian systems. Anat Rec B New Anat 2005;287:4-13.         [ Links ]

48. Morrison JI, Loof S, He P, Simon A. Salamander limb regeneration involves the activation of a multipotent skeletal muscle satellite cell population. J Cell Biol 2006;172:433-40.         [ Links ]

49. Otu HH, Naxerova K, Ho K, Can H, Nesbitt N, Libermann TA, et al. Restoration of liver mass after injury requires proliferative and not embryonic transcriptional patterns. J Biol Chem 2007;282:11197-204.         [ Links ]

50. Mastellos D, Lambris J D. Integrating innate immunity into a global systems context: the complement paradigm shift. En: Rigoutsos I, Stephanopoulus G, editors. Systems Biology. Vol. 2. Networks, models, and applications. Oxford: Oxford University Press; 2007. p. 169-92.         [ Links ]

51. Chu J, Sadler KC. New school in liver development: Lessons from zebrafish. Hepatology 2009;50:1656-63.         [ Links ]

52. Miyaoka YF, Ebato KF, Kato HF, Arakawa SF, Shimizu SF, Miyajima A. Hypertrophy and unconventional cell division of hepatocytes underlie liver regeneration. Curr Biol 2012;22:1166-75.         [ Links ]

53. Grisham JW. A morphologic study of deoxyribonucleic acid synthesis and cell proliferation in regenerating rat liver; autoradiography with thymidine-H3. Cancer Res 1962;22:842-9.         [ Links ]

54. Rock KL, Lai JJ, Kono H. Innate and adaptive immune responses to cell death. Immunol Rev 2011;243:191-205.         [ Links ]

55. Rock KL, Latz E, Ontiveros F, Kono H. The sterile inflammatory response. Annu Rev Immunol 2010;28:321-42.         [ Links ]

56. Barbalat R, Ewald SE, Mouchess ML, Barton GM. Nucleic acid recognition by the innate immune system. Annu Rev Immunol 2011;29:185-214.         [ Links ]

57. Lemasters JJ. Dying a thousand deaths: Redundant pathways from different organelles to apoptosis and necrosis. Gastroenterology 2005;129:351-60.         [ Links ]

58. Malhi H, Gores GJ, Lemasters JJ. Apoptosis and necrosis in the liver: A tale of two deaths? Hepatology 2006;43:S31-44.         [ Links ]

59. Kubes P, Mehal WZ. Sterile inflammation in the liver. Gastroenterology 2012;143:1158-72.         [ Links ]

60. Mohammed FF, Khokha R. Thinking outside the cell: Proteases regulate hepatocyte division. Trends Cell Biol 2005;15:555-63.         [ Links ]

61. Bohm F, Kohler UA, Speicher T, Werner S. Regulation of liver regeneration by growth factors and cytokines. EMBO Mol Med 2010;2:294-305.         [ Links ]

62. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010;140:805-20.         [ Links ]

63. Medzhitov R. Inflammation 2010: New adventures of an old flame. Cell 2010;140:771-6.         [ Links ]

64. Calfee CS, Matthay MA. Clinical immunology: Culprits with evolutionary ties. Nature 2010;464:41-2.         [ Links ]

65. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med 2009;361:1570-83.         [ Links ]

66. Mohammed FF, Pennington CJ, Kassiri Z, Rubin JS, Soloway PD, Ruther U, et al. Metalloproteinase inhibitor TIMP-1 affects hepatocyte cell cycle via HGF activation in murine liver regeneration. Hepatology 2005;41:857-67.         [ Links ]

67. Christophi C, Harun N, Fifis T. Liver regeneration and tumor stimulation -a review of cytokine and angiogenic factors. J Gastrointest Surg 2008;12:966-80.         [ Links ]

68. Malato Y, Naqvi S, Schurmann N, Ng R, Wang B, Zape J, et al. Fate tracing of mature hepatocytes in mouse liver homeostasis and regeneration. J Clin Invest 2011;121:4850-60.         [ Links ]

69. Morgan DO. The cell-cycle control system. En: Morgan DO, editor. The cell cycle: Principles of control. London: New Science Press; 2007. p. 27-55.         [ Links ]

70. Massague J. G1 cell-cycle control and cancer. Nature 2004;432:298-306.         [ Links ]

71. Michalopoulos GK. Liver regeneration after partial hepatectomy: Critical analysis of mechanistic dilemmas. Am J Pathol 2010;176:2-13.         [ Links ]

72. Kurinna S, Barton MC. Cascades of transcription regulation during liver regeneration. Int J Biochem Cell Biol 2011;43:189-97.         [ Links ]

73. Michalopoulos GK, DeFrances M. Liver regeneration. Adv Biochem Eng Biotechnol 2005;93:101-34.         [ Links ]

74. Duncan AW, Hickey RD, Paulk NK, Culberson AJ, Olson SB, Finegold MJ, et al. Ploidy reductions in murine fusion-derived hepatocytes. PLoS Genet 2009;5:e1000385.         [ Links ]

75. Duncan AW, Hanlon Newell AE, Smith L, Wilson EM, Olson SB, Thayer MJ, et al. Frequent aneuploidy among normal human hepatocytes. Gastroenterology 2012;142:25-8.         [ Links ]

76. Ding BS, Nolan DJ, Butler JM, James D, Babazadeh AO, Rosenwaks Z, et al. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature 2010;468:310-5.         [ Links ]

77. Maeno H, Ono T, Dhar DK, Sato T, Yamanoi A, Nagasue N. Expression of hypoxia inducible factor-1alpha during liver regeneration induced by partial hepatectomy in rats. Liver Int 2005;25:1002-9.         [ Links ]

78. Shimizu H, Miyazaki M, Wakabayashi Y, Mitsuhashi N, Kato A, Ito H, et al. Vascular endothelial growth factor secreted by replicating hepatocytes induces sinusoidal endothelial cell proliferation during regeneration after partial hepatectomy in rats. J Hepatol 2001;34:683-9.         [ Links ]

79. Drixler TA, Vogten MJ, Ritchie ED, van Vroonhoven TJ, Gebbink MF, Voest EE, et al. Liver regeneration is an angiogenesis-associated phenomenon. Ann Surg 2002;236:703,11; discussion 711-2.         [ Links ]

80. Drixler TA, Vogten JM, Gebbink MF, Carmeliet P, Voest EE, Borel Rinkes IH. Plasminogen mediates liver regeneration and angiogenesis after experimental partial hepatectomy. Br J Surg 2003;90:1384-90.         [ Links ]

81. Vollmar B, Menger MD. The hepatic microcirculation: Mechanistic contributions and therapeutic targets in liver injury and repair. Physiol Rev 2009;89:1269-339.         [ Links ]

82. Wu Y, Guo F, Liu J, Xiao X, Huang L, He D. Triple labeling with three thymidine analogs reveals a well-orchestrated regulation of hepatocyte proliferation during liver regeneration. Hepatol Res 2011;41:1230-9.         [ Links ]

83. Ferri D, Moro L, Mastrodonato M, Capuano F, Marra E, Liquori GE, et al. Ultrastructural zonal heterogeneity of hepatocytes and mitochondria within the hepatic acinus during liver regeneration after partial hepatectomy. Biol Cell 2005;97:277-88.         [ Links ]

84. Petersen BE, Zajac VF, Michalopoulos GK. Hepatic oval cell activation in response to injury following chemically induced periportal or pericentral damage in rats. Hepatology 1998;27:1030-8.         [ Links ]

85. Dabeva MD, Shafritz DA. Hepatic stem cells and liver repopulation. Semin Liver Dis 2003;23:349-62.         [ Links ]

86. Roskams TA, Libbrecht L, Desmet VJ. Progenitor cells in diseased human liver. Semin Liver Dis 2003;23:385-96.         [ Links ]

87. Van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol 2009;71:241-60.         [ Links ]

88. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663-76.         [ Links ]

89. Takahashi K, Okita K, Nakagawa M, Yamanaka S. Induction of pluripotent stem cells from fibroblast cultures. Nat Protoc 2007;2:3081-9.         [ Links ]

90. Cherry AB, Daley GQ. Reprogrammed cells for disease modeling and regenerative medicine. Annu Rev Med 2013;64:277-90.         [ Links ]

91. Bhave VS, Paranjpe S, Bowen WC, Donthamsetty S, Bell AW, Khillan JS, et al. Genes inducing iPS phenotype play a role in hepatocyte survival and proliferation in vitro and liver regeneration in vivo. Hepatology 2011;54:1360-70.         [ Links ]

92. Liu T, Wang Y, Tai G, Zhang S. Could co-transplantation of iPS cells derived hepatocytes and MSCs cure end-stage liver disease? Cell Biol Int 2009;33:1180-3.         [ Links ]

93. Locke JE, Shamblott MJ, Cameron AM. Stem cells and the liver: Clinical applications in transplantation. Adv Surg 2009;43:35-51.         [ Links ]

94. Sancho-Bru P, Najimi M, Caruso M, Pauwelyn K, Cantz T, Forbes S, et al. Stem and progenitor cells for liver repopulation: Can we standardise the process from bench to bedside? Gut 2009;58:594-603.         [ Links ]

95. Asgari S, Pournasr B, Salekdeh GH, Ghodsizadeh A, Ott M, Baharvand H. Induced pluripotent stem cells: A new era for hepatology. J Hepatol 2010;53:738-51.         [ Links ]

96. Diehl AM, Chute J. Underlying potential: cellular and molecular determinants of adult liver repair. J Clin Invest 2013;123:1858-60.         [ Links ]

97. DeLeve LD. Liver sinusoidal endothelial cells and liver regeneration. J Clin Invest 2013;123:1861-6.         [ Links ]

98. Yu Y, Fisher JE, Lillegard JB, Rodysill B, Amiot B, Nyberg SL. Cell therapies for liver diseases. Liver Transpl 2012;18:9-21.         [ Links ]

99. Furst G, Schulte am Esch J, Poll LW, Hosch SB, Fritz LB, Klein M, et al. Portal vein embolization and autologous CD133+ bone marrow stem cells for liver regeneration: Initial experience. Radiology 2007;243:171-9.         [ Links ]

100. Mertsching H, Schanz J, Steger V, Schandar M, Schenk M, Hansmann J, et al. Generation and transplantation of an autologous vascularized bioartificial human tissue. Transplantation 2009;88:203-10.         [ Links ]

101. Orlando G, Wood KJ, Stratta RJ, Yoo JJ, Atala A, Soker S. Regenerative medicine and organ transplantation: Past, present, and future. Transplantation 2011;91:1310-7.         [ Links ]

102. Zaret KS, Grompe M. Generation and regeneration of cells of the liver and pancreas. Science 2008;322:1490-4.         [ Links ]

103. Azuma H, Paulk N, Ranade A, Dorrell C, Al-Dhalimy M, Ellis E, et al. Robust expansion of human hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat Biotechnol 2007;25:903-10.         [ Links ]

104. Overturf K, Al-Dhalimy M, Finegold M, Grompe M. The repopulation potential of hepatocyte populations differing in size and prior mitotic expansion. Am J Pathol 1999;155:2135-43.         [ Links ]

105. Starzl TE, Fung J, Tzakis A, Todo S, Demetris AJ, Marino IR, et al. Baboon-to-human liver transplantation. Lancet 1993;341:65-71.         [ Links ]

106. Van Thiel DH, Gavaler JS, Kam I, Francavilla A, Polimeno L, Schade RR, et al. Rapid growth of an intact human liver transplanted into a recipient larger than the donor. Gastroenterology 1987;93:1414-9.         [ Links ]

107. Vauthey JN, Abdalla EK, Doherty DA, Gertsch P, Fenstermacher MJ, Loyer EM, et al. Body surface area and body weight predict total liver volume in Western adults. Liver Transpl 2002;8:233-40.         [ Links ]

108. Morgan DO. Control of cell proliferation and growth. In: Morgan DO, editor. The cell cycle: Principles of control. London: New Science Press; 2007. p. 195-225.         [ Links ]

109. Conlon I, Raff M. Size control in animal development. Cell 1999;96:235-44.         [ Links ]

110. Jorgensen P, Tyers M. How cells coordinate growth and division. Curr Biol 2004;14:R1014-27.         [ Links ]

111. Duncan AW, Taylor MH, Hickey RD, Hanlon Newell AE, Lenzi ML, Olson SB, et al. The ploidy conveyor of mature hepatocytes as a source of genetic variation. Nature 2010;467:707-10.         [ Links ]

112. Gupta S. Hepatic polyploidy and liver growth control. Semin Cancer Biol 2000;10:161-71.         [ Links ]

113. Lacroix B, Maddox AS. Cytokinesis, ploidy and aneuploidy. J Pathol 2012;226:338-51.         [ Links ]

114. Mitchison JM. Growth during the cell cycle. Int Rev Cytol 2003;226:165-258.         [ Links ]

115. Nurse P. Genetic control of cell size at cell division in yeast. Nature 1975;256:547-51.         [ Links ]

116. Gentric G, Celton-Morizur S, Desdouets C. Polyploidy and liver proliferation. Clin Res Hepatol Gastroenterol 2012;36:29-34.         [ Links ]

117. Kurinna S, Stratton SA, Coban Z, Schumacher JM, Grompe M, Duncan AW, et al. P53 Regulates a mitotic transcription program and determines ploidy in normal mouse liver. Hepatology 2013;57:2004-13.         [ Links ]

118. Minamishima YA, Nakayama K, Nakayama K. Recovery of liver mass without proliferation of hepatocytes after partial hepatectomy in Skp2-deficient mice. Cancer Res 2002;62:995-9.         [ Links ]

119. Wirth KG, Wutz G, Kudo NR, Desdouets C, Zetterberg A, Taghybeeglu S, et al. Separase: a universal trigger for sister chromatid disjunction but not chromosome cycle progression. J Cell Biol 2006;172:847-60.         [ Links ]

120. Edgar BA, Orr-Weaver TL. Endoreplication cell cycles: More for less. Cell 2001;105:297-306.         [ Links ]

121. Lu P, Prost S, Caldwell H, Tugwood JD, Betton GR, Harrison DJ. Microarray analysis of gene expression of mouse hepatocytes of different ploidy. Mamm Genome 2007;18:617-26.         [ Links ]

122. Ganem NJ, Godinho SA, Pellman D. A mechanism linking extra centrosomes to chromosomal instability. Nature 2009;460:278-82.         [ Links ]

123. Kamel IR, Erbay N, Warmbrand G, Kruskal JB, Pomfret EA, Raptopoulos V. Liver regeneration after living adult right lobe transplantation. Abdom Imaging 2003;28:53-7.         [ Links ]

124. Jin MB, Shimamura T, Taniguchi M, Nagasako Y, Suzuki T, Kamiyama T, et al. Liver regeneration in living-donor liver transplantation. Nihon Geka Gakkai Zasshi 2004;105:674-9.         [ Links ]

125. Humar A, Kosari K, Sielaff TD, Glessing B, Gomes M, Dietz C, et al. Liver regeneration after adult living donor and deceased donor split-liver transplants. Liver Transpl 2004;10:374-8.         [ Links ]

126. Mortensen KE, Conley LN, Hedegaard J, Kalstad T, Sorensen P, Bendixen C, et al. Regenerative response in the pig liver remnant varies with the degree of resection and rise in portal pressure. Am J Physiol Gastrointest Liver Physiol 2008;294:G819-30.         [ Links ]

127. Bucher NL, Swaffield MN. The rate of incorporation of labeled thymidine into the deoxyribonucleic acid of regenerating rat liver in relation to the amount of liver excised. Cancer Res 1964;24:1611-25.         [ Links ]

128. Weinbren K, Tarsh E. The mitotic response in the rat liver after different regenerative stimuli. Br J Exp Pathol 1964;45:475-80.         [ Links ]

129. Andersson U, Tracey KJ. Reflex principles of immunological homeostasis. Annu Rev Immunol 2012;30:313-35.         [ Links ]

130. Lin E, Calvano SE, Lowry SF. Inflammatory cytokines and cell response in surgery. Surgery 2000;127:117-26.         [ Links ]

131. Strey CW, Markiewski M, Mastellos D, Tudoran R, Spruce LA, Greenbaum LE, et al. The proinflammatory mediators C3a and C5a are essential for liver regeneration. J Exp Med 2003;198:913-23.         [ Links ]

132. DeAngelis RA, Markiewski MM, Lambris JD. Liver regeneration: A link to inflammation through complement. Adv Exp Med Biol 2006;586:17-34.         [ Links ]

133. Whitacre JM. Biological robustness: Paradigms, mechanisms, and systems principles. Front Genet 2012;3:67.         [ Links ]

134. Stelling J, Sauer U, Szallasi Z, Doyle FJ, 3rd, Doyle J. Robustness of cellular functions. Cell 2004;118:675-85.         [ Links ]

135. Kitano H. Cancer as a robust system: Implications for anticancer therapy. Nat Rev Cancer 2004;4:227-35.         [ Links ]

136. Corlu A, Loyer P. Regulation of the g1/s transition in hepatocytes: Involvement of the cyclin-dependent kinase cdk1 in the DNA replication. Int J Hepatol 2012;2012:689324.         [ Links ]

137. Tyson JJ, Chen KC, Novak B. Sniffers, buzzers, toggles and blinkers: Dynamics of regulatory and signaling pathways in the cell. Curr Opin Cell Biol 2003;15:221-31.         [ Links ]

138. Morgan DO. Cyclin-dependent kinases: engines, clocks, and microprocessors. Annu Rev Cell Dev Biol 1997;13:261-91.         [ Links ]

139. Albrecht JH, Mullany L K. Cell cycle control in the liver. En: Arias IM, Alter HJ, Boyer JL, Cohen DE, Fausto N, Shafritz DA, et al, editors. The liver: Biology and pathobiology. 5th ed. Oxford: Wiley & Sons; 2009. p. 1015-27.         [ Links ]

140. Pardee AB. G1 events and regulation of cell proliferation. Science 1989;246:603-8.         [ Links ]

141. Nurse P. Wee beasties. Nature 2004;432:557.         [ Links ]

142. Murray AW. Recycling the cell cycle: Cyclins revisited. Cell 2004;116:221-34.         [ Links ]

143. Stefater JA,3rd, Ren S, Lang RA, Duffield JS. Metchnikoff's policemen: Macrophages in development, homeostasis and regeneration. Trends Mol Med 2011;17:743-52.         [ Links ]

144. Malhi H, Gores GJ. Cellular and molecular mechanisms of liver injury. Gastroenterology 2008;134:1641-54.         [ Links ]

145. Malhi H, Guicciardi ME, Gores GJ. Hepatocyte death: A clear and present danger. Physiol Rev 2010;90:1165-94.         [ Links ]

146. Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ 2009;16:3-11.         [ Links ]

147. Guicciardi ME, Gores GJ. Apoptosis as a mechanism for liver disease progression. Semin Liver Dis 2010;30:402-10.         [ Links ]

148. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol 2008;8:279-89.         [ Links ]

149. Strey CW, Siegmund B, Rosenblum S, Marquez-Pinilla RM, Oppermann E, Huber-Lang M, et al. Complement and neutrophil function changes after liver resection in humans. World J Surg 2009;33:2635-43.         [ Links ]

150. Solis-Munoz P. Acute on chronic liver failure and prognostic factors: Time for reevaluation. Rev Esp Enferm Dig 2011;103:169-76.         [ Links ]

151. Helling TS. Liver failure following partial hepatectomy. HPB (Oxford) 2006;8:165-74.         [ Links ]

152. Hammond JS, Guha IN, Beckingham IJ, Lobo DN. Prediction, prevention and management of postresection liver failure. Br J Surg 2011;98:1188-200.         [ Links ]

153. Antoniades CG, Berry PA, Wendon JA, Vergani D. The importance of immune dysfunction in determining outcome in acute liver failure. J Hepatol 2008;49:845-61.         [ Links ]

154. Wasmuth HE, Kunz D, Yagmur E, Timmer-Stranghoner A, Vidacek D, Siewert E, et al. Patients with acute on chronic liver failure display "sepsis-like" immune paralysis. J Hepatol 2005;42:195-201.         [ Links ]

155. Van Golen RF, Van Gulik TM, Heger M. The sterile immune response during hepatic ischemia/reperfusion. Cytokine Growth Factor Rev 2012;23:69-84.         [ Links ]

156. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol 1994;12:991-1045.         [ Links ]

157. Matzinger P. An innate sense of danger. Semin Immunol 1998;10:399-415.         [ Links ]

158. Matzinger P. The danger model: A renewed sense of self. Science 2002;296:301-5.         [ Links ]

159. Mollen KP, Anand RJ, Tsung A, Prince JM, Levy RM, Billiar TR. Emerging paradigm: Toll-like receptor 4-sentinel for the detection of tissue damage. Shock 2006;26:430-7.         [ Links ]

160. Schwabe RF, Seki E, Brenner DA. Toll-like receptor signaling in the liver. Gastroenterology 2006;130:1886-900.         [ Links ]

161. Bianchi ME. DAMPs, PAMPs and alarmins: All we need to know about danger. J Leukoc Biol 2007;81:1-5.         [ Links ]

162. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004;4:499-511.         [ Links ]

163. Zimmermann HW, Trautwein C, Tacke F. Functional role of monocytes and macrophages for the inflammatory response in acute liver injury. Front Physiol 2012;3:56.         [ Links ]

164. Lamkanfi M, Dixit VM. Inflammasomes: Guardians of cytosolic sanctity. Immunol Rev 2009;227:95-105.         [ Links ]

165. Leemans JC, Cassel SL, Sutterwala FS. Sensing damage by the NLRP3 inflammasome. Immunol Rev 2011;243:152-62.         [ Links ]

166. Trauner M, Halilbasic E. Nuclear receptors as new perspective for the management of liver diseases. Gastroenterology 2011;140:1120-1125.e1-12.         [ Links ]

167. Medzhitov R, Schneider DS, Soares MP. Disease tolerance as a defense strategy. Science 2012;335:936-41.         [ Links ]

168. Basta G, Navarra T, De Simone P, Del Turco S, Gastaldelli A, Filipponi F. What is the role of the receptor for advanced glycation end products-ligand axis in liver injury? Liver Transpl 2011;17:633-40.         [ Links ]

169. Rovere-Querini P, Capobianco A, Scaffidi P, Valentinis B, Catalanotti F, Giazzon M, et al. HMGB1 is an endogenous immune adjuvant released by necrotic cells. EMBO Rep 2004;5:825-30.         [ Links ]

170. DeAngelis RA, Markiewski MM, Kourtzelis I, Rafail S, Syriga M, Sandor A, et al. A complement-IL-4 regulatory circuit controls liver regeneration. J Immunol 2012;188:641-8.         [ Links ]

171. Kohl J. The role of complement in danger sensing and transmission. Immunol Res 2006;34:157-76.         [ Links ]

172. Markiewski MM, DeAngelis RA, Strey CW, Foukas PG, Gerard C, Gerard N, et al. The regulation of liver cell survival by complement. J Immunol 2009;182:5412-8.         [ Links ]

173. Markiewski MM, Lambris JD. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am J Pathol 2007;171:715-27.         [ Links ]

174. Leslie M. Immunology. The new view of complement. Science 2012;337:1034-7.         [ Links ]

175. Matzinger P, Kamala T. Tissue-based class control: The other side of tolerance. Nat Rev Immunol 2011;11:221-30.         [ Links ]

176. Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol 2010;10:131-44.         [ Links ]

177. Seki E, Tsutsui H, Iimuro Y, Naka T, Son G, Akira S, et al. Contribution of Toll-like receptor/myeloid differentiation factor 88 signaling to murine liver regeneration. Hepatology 2005;41:443-50.         [ Links ]

178. Seki E, Schnabl B. Role of innate immunity and the microbiota in liver fibrosis: Crosstalk between the liver and gut. J Physiol 2012;590:447-58.         [ Links ]

179. Seki E, Brenner DA. Toll-like receptors and adaptor molecules in liver disease: Update. Hepatology 2008;48:322-35.         [ Links ]

180. Huang W, Ma K, Zhang J, Qatanani M, Cuvillier J, Liu J, et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 2006;312:233-6.         [ Links ]

181. Cornell RP, Liljequist BL, Bartizal KF. Depressed liver regeneration after partial hepatectomy of germ-free, athymic and lipopolysaccharide-resistant mice. Hepatology 1990;11:916-22.         [ Links ]

182. Cornell RP. Restriction of gut-derived endotoxin impairs DNA synthesis for liver regeneration. Am J Physiol 1985;249:R563-9.         [ Links ]

183. Cornell RP. Gut-derived endotoxin elicits hepatotrophic factor secretion for liver regeneration. Am J Physiol 1985;249:R551-62.         [ Links ]

184. Kim TH, Mars WM, Stolz DB, Petersen BE, Michalopoulos GK. Extracellular matrix remodeling at the early stages of liver regeneration in the rat. Hepatology 1997;26:896-904.         [ Links ]

185. Arthur MJ. Matrix degradation in liver: A role in injury and repair. Hepatology 1997;26:1069-71.         [ Links ]

186. Diehl AM. Cytokine regulation of liver injury and repair. Immunol Rev 2000;174:160-71.         [ Links ]

187. Van Sweringen HL, Sakai N, Tevar AD, Burns JM, Edwards MJ, Lentsch AB. CXC chemokine signaling in the liver: Impact on repair and regeneration. Hepatology 2011;54:1445-53.         [ Links ]

188. Hancock JT. Extracellular signals: hormones, cytokines and growth factors. En: Hancock JT, editor. Cell signalling. 3rd ed. Oxford: Oxford University Press; 2010. p. 55-77.         [ Links ]

189. Morello D, Fitzgerald MJ, Babinet C, Fausto N. c-myc, c-fos, and c-jun regulation in the regenerating livers of normal and H-2K/c-myc transgenic mice. Mol Cell Biol 1990;10:3185-93.         [ Links ]

190. Steer CJ. Liver regeneration. FASEB J 1995;9:1396-400.         [ Links ]

191. Li JW, Wang GP, Fan JY, Chang CF, Xu CS. Eight paths of ERK1/2 signalling pathway regulating hepatocyte proliferation in rat liver regeneration. J Genet 2011;90:435-42.         [ Links ]

192. Terui K, Ozaki M. The role of STAT3 in liver regeneration. Drugs Today (Barc) 2005;41:461-9.         [ Links ]

193. Selzner N, Selzner M, Odermatt B, Tian Y, Van Rooijen N, Clavien PA. ICAM-1 triggers liver regeneration through leukocyte recruitment and Kupffer cell-dependent release of TNF-alpha/IL-6 in mice. Gastroenterology 2003;124:692-700.         [ Links ]

194. Levy DE, Lee CK. What does Stat3 do? J Clin Invest 2002;109:1143-8.         [ Links ]

195. Li W, Liang X, Kellendonk C, Poli V, Taub R. STAT3 contributes to the mitogenic response of hepatocytes during liver regeneration. J Biol Chem 2002;277:28411-7.         [ Links ]

196. Malato Y, Sander LE, Liedtke C, Al-Masaoudi M, Tacke F, Trautwein C, et al. Hepatocyte-specific inhibitor-of-kappaB-kinase deletion triggers the innate immune response and promotes earlier cell proliferation during liver regeneration. Hepatology 2008;47:2036-50.         [ Links ]

197. Borowiak M, Garratt AN, Wustefeld T, Strehle M, Trautwein C, Birchmeier C. Met provides essential signals for liver regeneration. Proc Natl Acad Sci U S A 2004;101:10608-13.         [ Links ]

198. Taub R, Greenbaum LE, Peng Y. Transcriptional regulatory signals define cytokine-dependent and -independent pathways in liver regeneration. Semin Liver Dis 1999;19:117-27.         [ Links ]

199. Mastellos D, Papadimitriou JC, Franchini S, Tsonis PA, Lambris JD. A novel role of complement: Mice deficient in the fifth component of complement (C5) exhibit impaired liver regeneration. J Immunol 2001;166:2479-86.         [ Links ]

200. Aslam R, Speck ER, Kim M, Crow AR, Bang KW, Nestel FP, et al. Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo. Blood 2006;107:637-41.         [ Links ]

201. Semple JW, Italiano JE,Jr, Freedman J. Platelets and the immune continuum. Nat Rev Immunol 2011;11:264-74.         [ Links ]

202. Nowatari T, Fukunaga K, Ohkohchi N. Regulation of signal transduction and role of platelets in liver regeneration. Int J Hepatol 2012;2012:542479.         [ Links ]

203. Nurden AT. Platelets and tissue remodeling: Extending the role of the blood clotting system. Endocrinology 2007;148:3053-5.         [ Links ]

204. Tomiyama K, Miyazaki M, Nukui M, Takaishi M, Nakao A, Shimizu N, et al. Limited contribution of cells of intact extrahepatic tissue origin to hepatocyte regeneration in transplanted rat liver. Transplantation 2007;83:624-30.         [ Links ]

205. Alkozai EM, Nijsten MW, de Jong KP, de Boer MT, Peeters PM, Slooff MJ, et al. Immediate postoperative low platelet count is associated with delayed liver function recovery after partial liver resection. Ann Surg 2010;251:300-6.         [ Links ]

206. Lisman T, Porte RJ. The role of platelets in liver inflammation and regeneration. Semin Thromb Hemost 2010;36:170-4.         [ Links ]

207. Lesurtel M, Graf R, Aleil B, Walther DJ, Tian Y, Jochum W, et al. Platelet-derived serotonin mediates liver regeneration. Science 2006;312:104-7.         [ Links ]

208. Tian Y, Graf R, El-Badry AM, Lesurtel M, Furrer K, Moritz W, et al. Activation of serotonin receptor-2B rescues small-for-size liver graft failure in mice. Hepatology 2011;53:253-62.         [ Links ]

209. Matondo RB, Punt C, Homberg J, Toussaint MJ, Kisjes R, Korporaal SJ, et al. Deletion of the serotonin transporter in rats disturbs serotonin homeostasis without impairing liver regeneration. Am J Physiol Gastrointest Liver Physiol 2009;296:G963-8.         [ Links ]

210. Moolten FL, Bucher NL. Regeneration of rat liver: Transfer of humoral agent by cross circulation. Science 1967;158:272-4.         [ Links ]

211. Michalopoulos G, Cianciulli HD, Novotny AR, Kligerman AD, Strom SC, Jirtle RL. Liver regeneration studies with rat hepatocytes in primary culture. Cancer Res 1982;42:4673-82.         [ Links ]

212. Jirtle RL, Michalopoulos G. Effects of partial hepatectomy on transplanted hepatocytes. Cancer Res 1982;42:3000-4.         [ Links ]

213. Ozaki K, Leonard WJ. Cytokine and cytokine receptor pleiotropy and redundancy. J Biol Chem 2002;277:29355-8.         [ Links ]

214. Glotzbach JP, Wong VW, Gurtner GC, Longaker MT. Regenerative medicine. Curr Probl Surg 2011;48:148-212.         [ Links ]

215. Pennica D, King KL, Shaw KJ, Luis E, Rullamas J, Luoh SM, et al. Expression cloning of cardiotrophin 1, a cytokine that induces cardiac myocyte hypertrophy. Proc Natl Acad Sci U S A 1995;92:1142-6.         [ Links ]

216. Bustos M, Beraza N, Lasarte JJ, Baixeras E, Alzuguren P, Bordet T, et al. Protection against liver damage by cardiotrophin-1: A hepatocyte survival factor up-regulated in the regenerating liver in rats. Gastroenterology 2003;125:192-201.         [ Links ]

217. Bode JG, Heinrich P C. Interleukin-6 signaling during the acute-phase response of the liver. En: Arias IM, Boyer JL, Chisari FV, Fausto N, Schachter D, Shafritz DA, editors. The liver biology and pathobiology. 4th ed. Philadelphia: Lippincott Williams & Wilkins; 2001. p. 565-80.         [ Links ]

218. Szabo G, Romics L, Jr, Frendl G. Liver in sepsis and systemic inflammatory response syndrome. Clin Liver Dis 2002;6:1045,66, x.         [ Links ]

219. Streetz KL, Wustefeld T, Klein C, Manns MP, Trautwein C. Mediators of inflammation and acute phase response in the liver. Cell Mol Biol (Noisy-le-grand) 2001;47:661-73.         [ Links ]

220. Trautwein C, Boker K, Manns MP. Hepatocyte and immune system: Acute phase reaction as a contribution to early defence mechanisms. Gut 1994;35:1163-6.         [ Links ]

221. Zhang X, Mosser DM. Macrophage activation by endogenous danger signals. J Pathol 2008;214:161-78.         [ Links ]

222. Kishimoto T, Taga T, Akira S. Cytokine signal transduction. Cell 1994;76:253-62.         [ Links ]

223. Lutticken C, Wegenka UM, Yuan J, Buschmann J, Schindler C, Ziemiecki A, et al. Association of transcription factor APRF and protein kinase Jak1 with the interleukin-6 signal transducer gp130. Science 1994;263:89-92.         [ Links ]

224. Matsuda T, Hirano T. Association of p72 tyrosine kinase with Stat factors and its activation by interleukin-3, interleukin-6, and granulocyte colony-stimulating factor. Blood 1994;83:3457-61.         [ Links ]

225. Cressman DE, Greenbaum LE, DeAngelis RA, Ciliberto G, Furth EE, Poli V, et al. Liver failure and defective hepatocyte regeneration in interleukin-6-deficient mice. Science 1996;274:1379-83.         [ Links ]

226. Jin X, Zhang Z, Beer-Stolz D, Zimmers TA, Koniaris LG. Interleukin-6 inhibits oxidative injury and necrosis after extreme liver resection. Hepatology 2007;46:802-12.         [ Links ]

227. Iniguez M, Berasain C, Martinez-Anso E, Bustos M, Fortes P, Pennica D, et al. Cardiotrophin-1 defends the liver against ischemia-reperfusion injury and mediates the protective effect of ischemic preconditioning. J Exp Med 2006;203:2809-15.         [ Links ]

228. Oyama T, Sadamori H, Matsukawa H, Murata H, Umeda Y, Watanabe Y, et al. Small liver graft regenerates through immediate increase of HGF and IL-6--possible involvement of sinusoidal tensile/shear stress in small liver graft. Hepatogastroenterology 2007;54:2078-83.         [ Links ]

229. Blindenbacher A, Wang X, Langer I, Savino R, Terracciano L, Heim MH. Interleukin 6 is important for survival after partial hepatectomy in mice. Hepatology 2003;38:674-82.         [ Links ]

230. Beraza N, Marques JM, Martinez-Anso E, Iniguez M, Prieto J, Bustos M. Interplay among cardiotrophin-1, prostaglandins, and vascular endothelial growth factor in rat liver regeneration. Hepatology 2005;41:460-9.         [ Links ]

231. Tunon MJ, San Miguel B, Crespo I, Riezu-Boj JI, Larrea E, Alvarez M, et al. Cardiotrophin-1 promotes a high survival rate in rabbits with lethal fulminant hepatitis of viral origin. J Virol 2011;85:13124-32.         [ Links ]

232. Marques JM, Belza I, Holtmann B, Pennica D, Prieto J, Bustos M. Cardiotrophin-1 is an essential factor in the natural defense of the liver against apoptosis. Hepatology 2007;45:639-48.         [ Links ]

233. White P, Brestelli JE, Kaestner KH, Greenbaum LE. Identification of transcriptional networks during liver regeneration. J Biol Chem 2005;280:3715-22.         [ Links ]

234. Rodrigues CMP, Castro RE, Steer CJ. The role of bile acids in the modulation of apoptosis. Principles of Medical Biology 2004;15:119-45.         [ Links ]

235. Hulzebos CV, Renfurm L, Bandsma RH, Verkade HJ, Boer T, Boverhof R, et al. Measurement of parameters of cholic acid kinetics in plasma using a microscale stable isotope dilution technique: Application to rodents and humans. J Lipid Res 2001;42:1923-9.         [ Links ]

236. Palmeira CM, Rolo AP. Mitochondrially-mediated toxicity of bile acids. Toxicology 2004;203:1-15.         [ Links ]

237. Monte MJ, Martinez-Diez MC, El-Mir MY, Mendoza ME, Bravo P, Bachs O, et al. Changes in the pool of bile acids in hepatocyte nuclei during rat liver regeneration. J Hepatol 2002;36:534-42.         [ Links ]

238. Gravante G, Knowles T, Ong SL, Al-Taan O, Metcalfe M, Dennison AR, et al. Bile changes after liver surgery: Experimental and clinical lessons for future applications. Dig Surg 2010;27:450-60.         [ Links ]

239. Chiang JY. Regulation of bile acid synthesis: Pathways, nuclear receptors, and mechanisms. J Hepatol 2004;40:539-51.         [ Links ]

240. Martinez JD, Stratagoules ED, LaRue JM, Powell AA, Gause PR, Craven MT, et al. Different bile acids exhibit distinct biological effects: The tumor promoter deoxycholic acid induces apoptosis and the chemopreventive agent ursodeoxycholic acid inhibits cell proliferation. Nutr Cancer 1998;31:111-8.         [ Links ]

241. Fernandez-Barrena MG, Monte MJ, Latasa MU, Uriarte I, Vicente E, Chang HC, et al. Lack of Abcc3 expression impairs bile-acid induced liver growth and delays hepatic regeneration after partial hepatectomy in mice. J Hepatol 2012;56:367-73.         [ Links ]

242. Chen WD, Wang YD, Zhang L, Shiah S, Wang M, Yang F, et al. Farnesoid X receptor alleviates age-related proliferation defects in regenerating mouse livers by activating forkhead box m1b transcription. Hepatology 2010;51:953-62.         [ Links ]

243. Weymann A, Hartman E, Gazit V, Wang C, Glauber M, Turmelle Y, et al. P21 is required for dextrose-mediated inhibition of mouse liver regeneration. Hepatology 2009;50:207-15.         [ Links ]

244. Fisette A, Hassanain M, Metrakos P, Doi SA, Salman A, Schricker T, et al. High-dose insulin therapy reduces postoperative liver dysfunction and complications in liver resection patients through reduced apoptosis and altered inflammation. J Clin Endocrinol Metab 2012;97:217-26.         [ Links ]

245. Hassanain M, Metrakos P, Fisette A, Doi SA, Schricker T, Lattermann R, et al. Randomized clinical trial of the impact of insulin therapy on liver function in patients undergoing major liver resection. Br J Surg 2013;100:610-8.         [ Links ]

246. Costa RH, Kalinichenko VV, Holterman AX, Wang X. Transcription factors in liver development, differentiation, and regeneration. Hepatology 2003;38:1331-47.         [ Links ]

247. Wang X, Krupczak-Hollis K, Tan Y, Dennewitz MB, Adami GR, Costa RH. Increased hepatic Forkhead Box M1B (FoxM1B) levels in old-aged mice stimulated liver regeneration through diminished p27Kip1 protein levels and increased Cdc25B expression. J Biol Chem 2002;277:44310-6.         [ Links ]

248. Brezillon N, Lambert-Blot M, Morosan S, Couton D, Mitchell C, Kremsdorf D, et al. Transplanted hepatocytes over-expressing FoxM1B efficiently repopulate chronically injured mouse liver independent of donor age. Mol Ther 2007;15:1710-5.         [ Links ]

249. Marchioro TL, Porter KA, Brown BI, Otte JB, Starzl TE. The effect of partial portacaval transposition on the canine liver. Surgery 1967;61:723-32.         [ Links ]

250. Starzl TE, Porter KA, Francavilla A. The Eck fistula in animals and humans. Curr Probl Surg 1983;20:687-752.         [ Links ]

251. Starzl TE, Porter KA, Francavilla JA, Benichou J, Putnam CW. A hundred years of the hepatotrophic controversy. Ciba Found Symp 1977;(55):111-29.         [ Links ]

252. Francavilla A, Starzl TE, Porter K, Foglieni CS, Michalopoulos GK, Carrieri G, et al. Screening for candidate hepatic growth factors by selective portal infusion after canine Eck's fistula. Hepatology 1991;14:665-70.         [ Links ]

253. Gohda E, Tsubouchi H, Nakayama H, Hirono S, Sakiyama O, Takahashi K, et al. Purification and partial characterization of hepatocyte growth factor from plasma of a patient with fulminant hepatic failure. J Clin Invest 1988;81:414-9.         [ Links ]

254. Nakamura T, Nishizawa T, Hagiya M, Seki T, Shimonishi M, Sugimura A, et al. Molecular cloning and expression of human hepatocyte growth factor. Nature 1989;342:440-3.         [ Links ]

255. Nakamura T, Sakai K, Nakamura T, Matsumoto K. Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol 2011;26 Suppl 1:188-202.         [ Links ]

256. Michalopoulos G, Houck KA, Dolan ML, Leutteke NC. Control of hepatocyte replication by two serum factors. Cancer Res 1984;44:4414-9.         [ Links ]

257. Noji S, Tashiro K, Koyama E, Nohno T, Ohyama K, Taniguchi S, et al. Expression of hepatocyte growth factor gene in endothelial and Kupffer cells of damaged rat livers, as revealed by in situ hybridization. Biochem Biophys Res Commun 1990;173:42-7.         [ Links ]

258. Schirmacher P, Geerts A, Pietrangelo A, Dienes HP, Rogler CE. Hepatocyte growth factor/hepatopoietin A is expressed in fat-storing cells from rat liver but not myofibroblast-like cells derived from fat-storing cells. Hepatology 1992;15:5-11.         [ Links ]

259. Block GD, Locker J, Bowen WC, Petersen BE, Katyal S, Strom SC, et al. Population expansion, clonal growth, and specific differentiation patterns in primary cultures of hepatocytes induced by HGF/SF, EGF and TGF alpha in a chemically defined (HGM) medium. J Cell Biol 1996;132:1133-49.         [ Links ]

260. Rubin JS, Chan AM, Bottaro DP, Burgess WH, Taylor WG, Cech AC, et al. A broad-spectrum human lung fibroblast-derived mitogen is a variant of hepatocyte growth factor. Proc Natl Acad Sci U S A 1991;88:415-9.         [ Links ]

261. Hernandez J, Zarnegar R, Michalopoulos GK. Characterization of the effects of human placental HGF on rat hepatocytes. J Cell Physiol 1992;150:116-21.         [ Links ]

262. Mars WM, Liu ML, Kitson RP, Goldfarb RH, Gabauer MK, Michalopoulos GK. Immediate early detection of urokinase receptor after partial hepatectomy and its implications for initiation of liver regeneration. Hepatology 1995;21:1695-701.         [ Links ]

263. Hamanoue M, Kawaida K, Takao S, Shimazu H, Noji S, Matsumoto K, et al. Rapid and marked induction of hepatocyte growth factor during liver regeneration after ischemic or crush injury. Hepatology 1992;16:1485-92.         [ Links ]

264. Matsumoto K, Nakamura T. Emerging multipotent aspects of hepatocyte growth factor. J Biochem 1996;119:591-600.         [ Links ]

265. Matsumoto K, Nakamura T. Hepatocyte growth factor (HGF) as a tissue organizer for organogenesis and regeneration. Biochem Biophys Res Commun 1997;239:639-44.         [ Links ]

266. Benvenuti S, Comoglio PM. The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol 2007;213:316-25.         [ Links ]

267. Comoglio PM. Pathway specificity for Met signalling. Nat Cell Biol 2001;3:E161-2.         [ Links ]

268. Stolz DB, Mars WM, Petersen BE, Kim TH, Michalopoulos GK. Growth factor signal transduction immediately after two-thirds partial hepatectomy in the rat. Cancer Res 1999;59:3954-60.         [ Links ]

269. Lindroos PM, Zarnegar R, Michalopoulos GK. Hepatocyte growth factor (hepatopoietin A) rapidly increases in plasma before DNA synthesis and liver regeneration stimulated by partial hepatectomy and carbon tetrachloride administration. Hepatology 1991;13:743-50.         [ Links ]

270. Schmidt C, Bladt F, Goedecke S, Brinkmann V, Zschiesche W, Sharpe M, et al. Scatter factor/hepatocyte growth factor is essential for liver development. Nature 1995;373:699-702.         [ Links ]

271. Huh CG, Factor VM, Sanchez A, Uchida K, Conner EA, Thorgeirsson SS. Hepatocyte growth factor/c-met signaling pathway is required for efficient liver regeneration and repair. Proc Natl Acad Sci U S A 2004;101:4477-82.         [ Links ]

272. Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 2000;103:211-25.         [ Links ]

273. Sibilia M, Wagner EF. Strain-dependent epithelial defects in mice lacking the EGF receptor. Science 1995;269:234-8.         [ Links ]

274. Natarajan A, Wagner B, Sibilia M. The EGF receptor is required for efficient liver regeneration. Proc Natl Acad Sci U S A 2007;104:17081-6.         [ Links ]

275. Kiso S, Kawata S, Tamura S, Inui Y, Yoshida Y, Sawai Y, et al. Liver regeneration in heparin-binding EGF-like growth factor transgenic mice after partial hepatectomy. Gastroenterology 2003;124:701-7.         [ Links ]

276. Berasain C, Garcia-Trevijano ER, Castillo J, Erroba E, Lee DC, Prieto J, et al. Amphiregulin: An early trigger of liver regeneration in mice. Gastroenterology 2005;128:424-32.         [ Links ]

277. Loyer P, Cariou S, Glaise D, Bilodeau M, Baffet G, Guguen-Guillouzo C. Growth factor dependence of progression through G1 and S phases of adult rat hepatocytes in vitro. Evidence of a mitogen restriction point in mid-late G1. J Biol Chem 1996;271:11484-92.         [ Links ]

278. Loyer P, Corlu A, Desdouets C. Regulation of the hepatocyte cell cycle: Signaling pathways and protein kinases. Int J Hepatol 2012;2012:592354.         [ Links ]

279. Russell WE, Kaufmann WK, Sitaric S, Luetteke NC, Lee DC. Liver regeneration and hepatocarcinogenesis in transforming growth factor-alpha-targeted mice. Mol Carcinog 1996;15:183-9.         [ Links ]

280. Webber EM, Wu JC, Wang L, Merlino G, Fausto N. Overexpression of transforming growth factor-alpha causes liver enlargement and increased hepatocyte proliferation in transgenic mice. Am J Pathol 1994;145:398-408.         [ Links ]

281. Russell WE, Dempsey PJ, Sitaric S, Peck AJ, Coffey RJ, Jr. Transforming growth factor-alpha (TGF alpha) concentrations increase in regenerating rat liver: Evidence for a delayed accumulation of mature TGF alpha. Endocrinology 1993;133:1731-8.         [ Links ]

282. Campbell JS, Prichard L, Schaper F, Schmitz J, Stephenson-Famy A, Rosenfeld ME, et al. Expression of suppressors of cytokine signaling during liver regeneration. J Clin Invest 2001;107:1285-92.         [ Links ]

283. Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003;113:685-700.         [ Links ]

284. Moustakas A, Heldin CH. The regulation of TGFbeta signal transduction. Development 2009;136:3699-714.         [ Links ]

285. Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: Differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem 2007;101:9-33.         [ Links ]

286. Chari RS, Price DT, Sue SR, Meyers WC, Jirtle RL. Down-regulation of transforming growth factor beta receptor type I, II, and III during liver regeneration. Am J Surg 1995;169:126,31; discussion 131-2.         [ Links ]

287. Macias-Silva M, Li W, Leu JI, Crissey MA, Taub R. Up-regulated transcriptional repressors SnoN and Ski bind Smad proteins to antagonize transforming growth factor-beta signals during liver regeneration. J Biol Chem 2002;277:28483-90.         [ Links ]

288. Nishikawa Y, Wang M, Carr BI. 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 1998;176:612-23.         [ Links ]

289. Russell WE, Coffey RJ,Jr, Ouellette AJ, Moses HL. Type beta transforming growth factor reversibly inhibits the early proliferative response to partial hepatectomy in the rat. Proc Natl Acad Sci U S A 1988;85:5126-30.         [ Links ]

290. Bouzahzah B, Fu M, Iavarone A, Factor VM, Thorgeirsson SS, Pestell RG. Transforming growth factor-beta1 recruits histone deacetylase 1 to a p130 repressor complex in transgenic mice in vivo. Cancer Res 2000;60:4531-7.         [ Links ]

291. Oe S, Lemmer ER, Conner EA, Factor VM, Leveen P, Larsson J, et al. Intact signaling by transforming growth factor beta is not required for termination of liver regeneration in mice. Hepatology 2004;40:1098-105.         [ Links ]

292. Kogure K, Zhang YQ, Maeshima A, Suzuki K, Kuwano H, Kojima I. The role of activin and transforming growth factor-beta in the regulation of organ mass in the rat liver. Hepatology 2000;31:916-21.         [ Links ]

293. Uygun BE, Soto-Gutierrez A, Yagi H, Izamis ML, Guzzardi MA, Shulman C, et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nat Med 2010;16:814-20.         [ Links ]

294. Uygun BE, Yarmush ML, Uygun K. Application of whole-organ tissue engineering in hepatology. Nat Rev Gastroenterol Hepatol 2012;9:738-44.         [ Links ]

295. Soto-Gutierrez A, Wertheim JA, Ott HC, Gilbert TW. Perspectives on whole-organ assembly: Moving toward transplantation on demand. J Clin Invest 2012;122:3817-23.         [ Links ]

296. Hammond JS, Gilbert TW, Howard D, Zaitoun A, Michalopoulos G, Shakesheff KM, et al. Scaffolds containing growth factors and extracellular matrix induce hepatocyte proliferation and cell migration in normal and regenerating rat liver. J Hepatol 2011;54:279-87.         [ Links ]

297. Badylak SF, Taylor D, Uygun K. Whole-organ tissue engineering: Decellularization and recellularization of three-dimensional matrix scaffolds. Annu Rev Biomed Eng 2011;13:27-53.         [ Links ]

298. Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S. The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology 2011;53:604-17.         [ Links ]

299. Gkretsi V, Bowen WC, Yang Y, Wu C, Michalopoulos GK. Integrin-linked kinase is involved in matrix-induced hepatocyte differentiation. Biochem Biophys Res Commun 2007;353:638-43.         [ Links ]

300. Gkretsi V, Apte U, Mars WM, Bowen WC, Luo JH, Yang Y, et al. Liver-specific ablation of integrin-linked kinase in mice results in abnormal histology, enhanced cell proliferation, and hepatomegaly. Hepatology 2008;48:1932-41.         [ Links ]

301. Apte U, Gkretsi V, Bowen WC, Mars WM, Luo JH, Donthamsetty S, et al. Enhanced liver regeneration following changes induced by hepatocyte-specific genetic ablation of integrin-linked kinase. Hepatology 2009;50:844-51.         [ Links ]

302. Shafiei MS, Rockey DC. The role of integrin-linked kinase in liver wound healing. J Biol Chem 2006;281:24863-72.         [ Links ]

303. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP, Ott HC. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med 2013;19:646-51.         [ Links ]

304. Song JJ, Kim SS, Liu Z, Madsen JC, Mathisen DJ, Vacanti JP, et al. Enhanced in vivo function of bioartificial lungs in rats. Ann Thorac Surg 2011;92:998,1005; discussion 1005-6.         [ Links ]

305. Bakkenist CJ, Kastan MB. Initiating cellular stress responses. Cell 2004;118:9-17.         [ Links ]

306. Ben-Porath I, Weinberg RA. When cells get stressed: An integrative view of cellular senescence. J Clin Invest 2004;113:8-13.         [ Links ]

307. Morgan DO. The DNA damage response. In: Morgan DO, editor. The cell cycle: Principles of control. London: New Science Press; 2007. p. 227-45.         [ Links ]

308. Campisi J. Senescent cells, tumor suppression, and organismal aging: Good citizens, bad neighbors. Cell 2005;120:513-22.         [ Links ]

309. Weinberg RA. P53 and apoptosis: master guardian and executioner. In: Weinberg RA, editor. The biology of cancer. New York: Garland Science; 2007. p. 307-56.         [ Links ]

310. Vousden KH, Lu X. Live or let die: the cell's response to p53. Nat Rev Cancer 2002;2:594-604.         [ Links ]

311. Levine AJ. P53, the Cellular Gatekeeper for Growth and Division. Cell 1997;88:323-31.         [ Links ]

312. Efeyan A, Serrano M. P53: Guardian of the genome and policeman of the oncogenes. Cell Cycle 2007;6:1006-10.         [ Links ]

313. Li CC, Chu HY, Yang CW, Chou CK, Tsai TF. Aurora-A overexpression in mouse liver causes p53-dependent premitotic arrest during liver regeneration. Mol Cancer Res 2009;7:678-88.         [ Links ]

314. Maddocks OD, Vousden KH. Metabolic regulation by p53. J Mol Med (Berl) 2011;89:237-45.         [ Links ]

315. 413.Fededa JP, Gerlich DW. Molecular control of animal cell cytokinesis. Nat Cell Biol 2012;14:440-7.         [ Links ]

316. Gallagher SJ, Kefford RF, Rizos H. The ARF tumour suppressor. Int J Biochem Cell Biol 2006;38:1637-41.         [ Links ]

317. Wadhwa R, Sugihara T, Taira K, Kaul SC. The ARF-p53 senescence pathway in mouse and human cells. Histol Histopathol 2004;19:311-6.         [ Links ]

318. Zhou J, Schmid T, Schnitzer S, Brune B. Tumor hypoxia and cancer progression. Cancer Lett 2006;237:10-21.         [ Links ]

319. Demetris AJ, Seaberg EC, Wennerberg A, Ionellie J, Michalopoulos G. Ductular reaction after submassive necrosis in humans. Special emphasis on analysis of ductular hepatocytes. Am J Pathol 1996;149:439-48.         [ Links ]

320. Stravitz RT, Kramer D J. Acute liver failure. En: Boyer TD, Manns MP, Sanyal AJ, editores. Zakim and Boyer's hepatology: a textbook of liver disease. 6th ed. Philadelphia: Saunders Elsevier; 2012. p. 327-51.         [ Links ]

321. Oda M, Yokomori H, Han JY. Regulatory mechanisms of hepatic microcirculatory hemodynamics: hepatic arterial system. Clin Hemorheol Microcirc 2006;34:11-26.         [ Links ]

322. Ninomiya M, Shirabe K, Terashi T, Ijichi H, Yonemura Y, Harada N, et al. Deceleration of regenerative response improves the outcome of rat with massive hepatectomy. Am J Transplant 2010;10:1580-7.         [ Links ]

323. Markiewski MM, DeAngelis RA, Lambris JD. Liver inflammation and regeneration: Two distinct biological phenomena or parallel pathophysiologic processes? Mol Immunol 2006;43:45-56.         [ Links ]

324. Clavien PA, Oberkofler CE, Raptis DA, Lehmann K, Rickenbacher A, El-Badry AM. What is critical for liver surgery and partial liver transplantation: Size or quality? Hepatology 2010;52:715-29.         [ Links ]

325. Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 2013;499:481-4.         [ Links ]

326. Ordonez MP, Goldstein LS. Using human-induced pluripotent stem cells to model monogenic metabolic disorders of the liver. Semin Liver Dis 2012;32:298-306.         [ Links ]

327. Beginner´s Traps. In: Ramón y Cajal S, editor. Advice for a young investigator. Cambridge, Massachussetts: Massachussetts Institute of Technology; 1999. p. 9-27.         [ Links ]

 

 

Correspondence:
Javier A.-Cienfuegos.
Department of General Surgery.
Clínica Universidad de Navarra.
Avda. Pío XII, no 36.
31008 Pamplona, Navarra. Spain
e-mail: fjacien@unav.es

Received: 19-07-2013
Accepted: 12-11-2013

Creative Commons License Todo el contenido de esta revista, excepto dónde está identificado, está bajo una Licencia Creative Commons