Skip to main content
Erschienen in: Hepatology International 4/2020

Open Access 22.06.2020 | Review Article

Cell cycle regulation in NAFLD: when imbalanced metabolism limits cell division

verfasst von: Matias J. Caldez, Mikael Bjorklund, Philipp Kaldis

Erschienen in: Hepatology International | Ausgabe 4/2020

Abstract

Cell division is essential for organismal growth and tissue homeostasis. It is exceptionally significant in tissues chronically exposed to intrinsic and external damage, like the liver. After decades of studying the regulation of cell cycle by extracellular signals, there are still gaps in our knowledge on how these two interact with metabolic pathways in vivo. Studying the cross-talk of these pathways has direct clinical implications as defects in cell division, signaling pathways, and metabolic homeostasis are frequently observed in liver diseases. In this review, we will focus on recent reports which describe various functions of cell cycle regulators in hepatic homeostasis. We will describe the interplay between the cell cycle and metabolism during liver regeneration after acute and chronic damage. We will focus our attention on non-alcoholic fatty liver disease, especially non-alcoholic steatohepatitis. The global incidence of non-alcoholic fatty liver disease is increasing exponentially. Therefore, understanding the interplay between cell cycle regulators and metabolism may lead to the discovery of novel therapeutic targets amenable to intervention.
Hinweise
Lead Contact: Philipp Kaldis.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The liver is the largest solid organ in the mammalian body. It fulfills essential roles in the gastrointestinal tract, controlling metabolism, detoxification, digestion, and many other essential processes [1]. Due to its functions and the strategic location alongside the gastrointestinal tract, it is constantly exposed to intrinsic and external damage. To maintain functionality, the liver has retained an impressive capacity to regenerate after injury [2]. The mechanisms underlying liver regeneration are classified into acute or chronic damage depending on nature, timing, and duration of the injury [3]. The critical difference is that regeneration after acute damage represents a synchronized physiological process [4]. In contrast, chronic damage is characterized by cycles of damage-regeneration similar to a wound healing response (Fig. 1). In this review, we will explore the interaction between metabolism and cell cycle in tissue regeneration after partial hepatectomy as a model of acute damage. Next, we will compare these processes in chronic damage, focusing especially in non-alcoholic fatty liver disease (NAFLD).
NAFLD represents the most frequent chronic liver disease in the world. It is characterized by the accumulation of lipid droplets in hepatic cells [5], combined with dysregulation of metabolism and cellular division [4]. Many questions remain open about the role of cell cycle and metabolic pathways during hepatic regeneration after acute and chronic damage. To resolve some of these questions, this review is divided into two parts: (1) in the first section, we will summarize recent data on various roles of cell cycle genes involved in hepatocyte division. Simultaneously, we will describe in detail the functions of metabolic genes during liver regeneration after hepatectomy as an example of acute liver damage (Fig. 1a). Then, (2) we will briefly outline data of studies on the regulation of the cell cycle in murine models of NAFLD as an example of chronic damage. Ultimately, we will propose potential nodes for targeting the interface between the cell cycle and metabolism to improve NAFLD treatment. Liver regeneration after hepatectomy is the standard method to study the synchronized regulation of cell cycle and metabolism in vivo. However, it is essential to highlight that the mechanism of tissue regeneration after resection is different from that of NAFLD.

The regulation of the cell cycle, signaling, and metabolism during acute hepatic damage and regeneration

Liver regeneration after acute injury strands for an adaptive response by which the liver compensates for the loss of functional tissue using cellular hypertrophy (increase in the size of hepatic cells) and/or hyperplasia ([2, 3], Fig. 1a). Two features render liver regeneration unique compared to repair and healing of other tissues. First, before injury, hepatocytes are in a quiescent state. However, when the liver is confronted with damage hepatocytes are able to re-enter the cell cycle despite being fully differentiated. Therefore, they may function as “facultative” stem cells (for more details on the role of liver stem cells in liver regeneration refer to [2]). Second, hepatic regeneration is well synchronized. Third, the rate of cellular division during regeneration is controlled by several factors. Growth factors from other organs including cytokines from an inflammatory response, metabolic signals, circadian rhythm, and blood pressure are among the most significant ones [2].
The best-described models to study liver regeneration after acute damage are (1) liver resection (partial hepatectomy [3]) and (2) toxicological damage [6]. The essential difference is that the latter induces parenchymatic necrosis, and hepatocyte proliferation is integrated into a complex wound healing response with fibrosis and inflammation [6]. In contrast, 2/3 liver resection activates cell division in the absence of any cellular lesion [3]. In the following discussion, the regenerative response of the liver to partial hepatectomy will be divided into three groups of processes, namely: (1) the activation of cellular signaling pathways; (2) regulation of cell cycle progression; and (3) remodeling of metabolism (Fig. 1a). While these events are occurring simultaneously, the proposed classification solely aims to facilitate the description. We will initially introduce general ideas about the regulation of these three processes independently. Subsequently, we will highlight potential nodes of interaction between the three groups of biological processes.

Regulation of cellular signaling during liver regeneration

Resection of 2/3 of the liver activates an acute phase response followed by a profound rearrangement of the entire cellular signaling network [7]. Based on the consecutive activation of a diverse group of transcription factors (TFs), the process of liver regeneration is conceptually divided into three phases: priming, progression, and termination (Fig. 2). The initiation phase is characterized by a fast activation of early-response genes [2] with a dramatic increase in blood pressure [8]. Collectively, these initiating events have been associated with the release of IL-6 and activation of the IL-6-gp130-Stat3 axis in Kupffer cells [9]. IL-6 from Kupffer cells activates the IL-6 receptor gp130 in hepatocytes which in turn will phosphorylate STAT3, causing increases in transcriptional activity (Fig. 2). Early response TFs promote the activation of a second group known as late-response genes that undergo de novo transcription and translation [2]. Combined with other TFs activated by growth factors like hepatocyte and epidermal growth factors (EGF, HGF), the IL-6-gp130 axis activates expression of key cell cycle genes [1015].
Cell cycle genes drive the progression phase until the onset of the termination phase [16]. Within 1–2 weeks after resection, any remaining cellular division will eventually cease in murine livers. As such, two weeks after resection, the liver will adjust to the initial pre-resection size and function while in other mammals the process may take longer time. The final stage of regeneration is characterized by the termination phase. Here, the liver-to-body weight ratio is carefully adjusted. The ratio is regulated by the rate of hepatic division and apoptosis until the liver reaches an adequate size and all normal physiological functions are restored [17, 18].

Regulation of the cell cycle: the role of hypertrophy and cellular division during regeneration

Another distinctive feature of liver regeneration is the dynamic alterations in cellular hypertrophy and division [19]. Normally, after hepatocytes are primed by injury, they temporarily increase in size with a subsequent activation of cell cycle genes and cellular division [17]. However, we and others have shown that an increase in hepatocyte size alone is sufficient to recover organ mass in the absence of cell division in mice. For example deletion of the cell cycle genes Skp2 or Cdk1 [2023] does not impair regeneration. In addition, early observations indicated that components of the mTOR-AKT signaling are essential to activate an alternative regenerative response to promote tissue recovery by compensatory cellular hypertrophy [19, 24, 25]. Nevertheless, we also observed that compensatory cellular hypertrophy is sufficient for mass recovery, but liver functions are reduced [26]. On top of that, our recent unpublished data suggest that in the long-term, increases in the size of hepatocytes may induce activation of an inflammatory response and fibrosis. In conclusion, in the short term cell hypertrophy may offer an alternative form of tissue regeneration, but may not be sufficient for recovery of liver functions.
During liver regeneration, hepatocytes display an equivalent pattern of expression of cell cycle genes comparable to that observed in cell culture models in vitro (for an exhaustive review on cell cycle regulators refer to Refs. [27, 28] and Fig. 2). For example, before liver resection, most genes involved in cell division like Cdk1 and cyclin B are downregulated. Instead genes involved in quiescence like p21Cip1/Waf1 and p27Kip1 are highly expressed [12, 2931]. During the initiation phase of liver regeneration, the expression of genes involved in G1 progression like cyclin D and Cdk4 is induced [29]. Finally, during the progression phase of regeneration most cell cycle genes are actively expressed [32, 33] (Fig. 3). However, there are several exceptions to this rule. For example, the murine cyclin B transcript is regulated at different time points during liver regeneration, with a peak of expression that coincides with the peak of DNA replication (S-phase, ≈ 36 h after resection) [34, 35]. Additionally, in regenerating livers cyclin A and cyclin B are observed in the cytoplasmic, nuclear, and membrane containing fractions [36, 37]. In the same way, Cdk1 is expressed as early as 30 h after hepatectomy, which is the period of DNA replication in regenerating hepatocytes [38]. These observations suggest that Cdk1 may display novel functions outside of mitosis, which is supported by recent data [26, 38] but was not observed in vitro models. Collectively, these results suggest that Cdk1 and its partners cyclin A and cyclin B, may possess non-canonical functions in the liver, different to the ones described in cell culture systems. We and others have shown that during liver regeneration Cdk1 controls cell size and cell division [39], cell metabolism [26], and DNA replication [38, 40]. However, little data is available on the partners cyclin A and cyclin B. Hence, further studies may be essential to understand the roles of these cyclins during liver regeneration.

Metabolic regulation during liver regeneration after acute liver damage

The liver is a vital center for homeostasis of glucose, lipid, and overall metabolism [41]. Therefore, acute tissue damage imposes significant stress to the general metabolic performance of the whole organ. Nevertheless, during liver regeneration after 2/3 liver resection, hepatocytes are able to remodel their metabolic network to promote regeneration while maintaining overall systemic metabolic balance. These early observations suggest that liver metabolism is robust. In this sense, metabolic pathways compensate for the loss of many individual enzymes, which is supported by mouse genetics [4245]. However, liver specific deletion of some enzymes including nicotinamide phosphoribosyl transferase (Nampt) and several members of the P450 cytochrome reductase family leads to lethality after partial hepatectomy [4648].
The changes in metabolic demands usually induce small rearrangements in metabolism through local mechanisms without substantial rewiring of the overall metabolic network [41]. However, 2/3 partial hepatectomy leads to both loss of tissue mass and glycogen, a major energy reservoir for systemic glucose and energy metabolism [49]. Major resection of glycogen in the liver, leads to a substantial reduction in blood glucose. This is particularly apparent during the initiation phase of liver regeneration when there is a significant increase in the oxidative status as a response to a whole body induction of catabolism [49]. Because glucose supply via glycogenolysis is limited, the energy required to promote liver regeneration may be partly acquired from alternative sources that are actively transported into hepatocyte, such as lipids [50]. In fact, deletion of the Caveolin1 (Cav1) gene induced hepatic failure during early phases of liver regeneration, indicating that lipid import into hepatocytes is essential to drive regeneration [51]. CAV1 is essential to transport lipids into hepatocytes. Therefore, these observations are in agreement with temporary hepatic steatosis being a key feature of the early stages of liver regeneration [52, 53]. Interestingly, physiological steatosis does not overlap with cell division during liver regeneration, which may imply that lipid metabolism and cell division are strictly temporally separated. However, these ideas need to be further tested.
During the termination phase of liver regeneration, hepatic gluconeogenic functions regain homeostatic levels, marked by a sharp increase in expression of Akt [44]. These events are followed by the activation of biosynthetic pathways to promote tissue homeostasis, accumulation of glycogen as well as normal glucose and lipid metabolic processes. Based on the main two primary sources of energy, we would like to propose a division of metabolic-related events during liver regeneration in two phases (1) oxidative and (2) biosynthetic (Fig. 4). The oxidation phase is distinguished by oxidative metabolism correlated to increases in cell size. This would last from the early minutes after liver resection to 30 h, when hepatic DNA synthesis starts to peak (Fig. 4). After that, the biosynthetic pathways will be active until metabolic homeostasis is reached.

Interactions between cell cycle regulators and metabolism during acute liver damage

Reduction in blood glucose represents one of the primary metabolic markers of hepatic regeneration [26]. The characteristic short-term hypoglycemia is required for adequate liver regeneration. This is supported by clinical data indicating that patients with hyperglycemia typically experience an adverse regenerative outcome [54]. Another cause for decreased concentration of glucose in serum is the reduced expression of metabolic enzymes during the priming and progression phases [17]. We have recently published similar findings in mice employing a combination of transcriptomic, metabolomic and advanced molecular imaging [26].
During cellular division CDK1 phosphorylates specific subunits of the mitochondrial oxidative phosphorylation complex I [55, 56]. In this sense, CDK1 may couple cell division and energy metabolism. In post-mitotic tissues, CDK1 regulates gluconeogenic pathways by phosphorylation of FOXO-1 [57] and glycolytic genes like PFKFB3 [58, 59]. Following this line of evidence, we published that ablation of CDK1 in hepatocytes led to a significant reduction in NADH concentration because mitochondrial function is suboptimal [60]. These observations are in agreement with recent reports suggesting that deletion of Nampt reduced hepatocyte proliferation during regeneration [47]. Collectively, these results suggest an interplay between the oxidative state of hepatocytes and CDK1. Deletion of Cdk1 in the liver led to a significant reduction in mitochondrial metabolic processes during S-phase [26]. A standard explanation for this is that DNA replication and oxidative metabolism are not compatible. Reactive oxygen species (ROS) generated as a side-product of oxidative metabolism would be detrimental when the maintenance of DNA fidelity is most critical [61, 62].

Cholesterol and the cell cycle

In the previous sections, we highlighted that hepatic accumulation of lipid droplets is associated with the initiation of regeneration and not cellular division. Therefore, we speculate that the presence of either lipid droplets or their components are incompatible with proliferation. A lipid droplet is composed by triglycerides, lipoproteins, cholesterol and other components [63]. Cholesterol represents the fundamental structural component and thus is essential for its formation. In mammals there are two main sources of cholesterol, (1) gastrointestinal digestion and (2) biosynthesis. Regardless of the source, cholesterol is stored and metabolized as esterified cholesterol in pericentral hepatocytes [64]. As mentioned earlier, during liver regeneration the metabolic capacity of hepatocytes is heavily reduced, and it is expected that cholesterol production may be hindered as well. In fact, our preliminary data show that concentration of serum total, free and esterified cholesterol are significantly reduced during liver regeneration (unpublished).
In mice, hepatic Srebp-1c is an essential gene for the transcriptional activation of enzymes involved in cholesterol metabolic processes. Deletion of Srebp-1c in hepatocytes led to increased accumulation of cholesterol [65], with no changes in the composition of other lipids. When mice without Srebp-1c were subjected to 2/3 partial hepatectomy, hepatocyte division was significantly enhanced in comparison to wild type mice [65]. This was in agreement with previous observations that showed reduced expression of CDK1 and less mitotic cells upon cholesterol starvation [66, 67]. Together, these studies further support the idea that there might be a relationship between cholesterol metabolism and cellular division. We have shown that reduced division rate of cells during liver regeneration, led to a significant reduction in expression of enzymes from cholesterol and mevalonate pathway, suggesting an interaction between the machinery that regulates cell cycle and that of cholesterol synthesis. This is further supported by experiments indicating that during cell cycle progression, cells regulate their lipid composition and localization [68]. We are currently dissecting the molecular mechanism by which CDK1 may regulate cholesterol synthesis in hepatocytes. Forthcoming work should address the association between the formation of lipid droplets during physiological steatosis and cellular division during liver regeneration independently.

The bidirectional interaction of metabolism and cell division during liver regeneration after chronic damage

NAFLD is characterized by a sustained insult to the parenchyma, with alternating cycles of damage and regeneration. The liver does not regenerate optimally in patients with NAFLD, NASH, and/or obese patients [69, 70]. Indeed, it has been proposed that steatosis in patients before liver resection is a risk factor for liver failure after hepatectomy [70] and causes postoperative complications [71]. Growing evidence has indicated that NAFLD is typically associated with increased synthesis of lipids derived from long-chain fatty acids, like palmitic and linoleic acids [72]. These macromolecules promote the characteristic inflammatory response through the production of lipotoxic molecules [72]. However, and more importantly, these bioactive lipid precursors usually activate lipid peroxidation, with a concomitant reduction in mitochondria function and increased oxidative stress by production of ROS [72]. In this context, cell cycle progression is impaired, with concomitant activation of apoptosis which further promotes inflammation and fibrosis. In contrast to a healthy liver, hepatocyte-dependent regeneration is diminished in the chronically damaged liver. In this section, the potential associations between cell division and metabolism will be discussed in two in models of diabetes, obesity, NAFLD, and NASH.

Hepatocyte cell cycle progression in genetic models of obesity and diabetes

NAFLD evolves from the response of altered (1) lipid transport into hepatocytes; (2) increased production of lipids from hepatocytes; as well as (3) reduced export and consumption of lipids [63]. In diabetes and obesity, NAFLD is accompanied by changes in insulin metabolism [5]. In hepatocytes, insulin promotes the biosynthesis of lipids by inhibiting fatty acid oxidation, with leptin being essential for the insulin response [73]. Murine models with mutations in either leptin or the leptin receptor gene led to body fat accumulation, and the onset of obesity and diabetes mellitus. Initial observations using these mouse models demonstrated impaired cell cycle progression of hepatocytes after liver regeneration. For example, hepatocytes from Zucker rats arrested in G1, with a concomitant lower expression of cyclin D1 after hepatectomy [74]. These initial results led to the hypothesis that accumulation of fat droplets may negatively regulate hepatic cell division and reduce cell proliferation. Besides, in ob/ob mice survival rate after liver resection was significantly also lower than in wild type. These mice do not produce leptin, and displayed significant hypoglycemia as well as impaired hepatocellular proliferation after liver resection [75]. These effects occur with no difference in serum concentration of pro-proliferative hormones during liver regeneration like TNFα, IL-6, and insulin. In a third model, fa/fa rats displayed elevated concentration of leptin in blood and a similar reduction in the survival after hepatectomy [76]. Nonetheless, leptin replacement restored TNFα and IL-6 release and induced cyclin D1, suggesting that leptin may play a central role in the interaction between cell cycle regulators and lipid metabolism [77]. A conflicting result arises from the fact that leptin supplementation improved hepatic division, but did not reduce the onset of liver failure [78, 79]. In addition, intraperitoneal injection of leptin in wild type mice with no obesity increased mitotic counts during liver regeneration [78]. Collectively, these results suggest that adipokines play a significant role in hepatocellular proliferation. Similar results were observed in a model of fatty liver in ALPPS rats [80]. In summary, leptin is essential for hepatic division and together with other factors may act in recovery after hepatectomy [81]. Subsequent research should address the role of adipokines like leptin in regulation of hepatic cell cycle genes.

Cell cycle progression in models of diet manipulation

To promote lipid overloading independently of a genetic mutation, many experiments provide mice with a high-fat diet (HFD). These models are more clinically-relevant compared to mice with mutations in enzymes. HFD leads to high calorie intake, body weight increase, liver steatosis and reduced regeneration caused by metabolic surplus [82, 83]. Liver regeneration in mice fed with HFD was accompanied by increased TNF-α potentially amplified by leptin which promoted increased levels of IκBα preventing NF-κB activation. Consequently, expression of NF-κB target genes, including cyclin D1 was reduced [82] (Fig. 5) leading to a decrease in proliferation of hepatocytes. Mice fed with HFD display a delayed incorporation of BrdU after surgery [84], indicating that steatosis not only inhibits cell division but also delays DNA replication. Expression of cyclin D1 and cyclin E2 are usually higher before partial hepatectomy in mice fed with HFD. However, there was no significant difference in expression between mice fed with HFD and Chow diets during liver regeneration [84]. In contrast, cyclin A2 and cyclin B1 are significantly reduced during liver regeneration in mice fed with HFD, further suggesting that cell division might be impaired [82]. In conclusion, these observations suggest that leptin-related steatosis due to HFD may regulate cell cycle progression differentially by downstream control of the inflammatory pathway NF-κB, which is supported by studies done in mice with lipodystrophy [85]. Lipid synthesis and degradation, as well as the correct localization of the corresponding enzymes are essential for cell division [68]. To the same extent, cell cycle effectors regulate directly or indirectly lipid synthesis in hepatocytes. To follow up on these topics, in the next section we will summarize current data on how cell cycle regulators control lipid synthesis. We will particularly focus on a novel axis of molecular signaling that we describe as the p21-CDK1-E2F1-lipid synthesis axis.

Lipid metabolism and cell cycle in NAFLD

NAFLD pathogenesis displays a broad phenotype ranging from cellular steatosis, characterized by micro and macrovesicles, to NASH with substantial inflammation and fibrosis. As mentioned above, an increasing amount of evidence indicates that the synthesis of a particular group of lipids in NAFLD hepatocytes may initiate the activation of inflammation. In recent years, several reports have suggested that the direct consequence of the chronic metabolic inflammation is the activation of senescence-associated secretory phenotype (SASP) [4] with a concomitant increase in expression of the senescence marker p21Cip1/Waf1 [86]. Cellular senescence is a biological process activated by an irreversible cell cycle arrest. Replicative senescence is associated with aging of tissues, mainly caused by shortening of the telomeres. In contrast, stress-induced premature senescence is caused by DNA damage caused by intra or extracellular factors such as oxidative stress. Either type of senescence leads to a DNA damage response, which inhibits cell cycle progression. In NAFLD, chronic metabolic surplus generates many of the molecules involved in cellular senescence, like ROS, with a concomitant change in gene expression. Several genome wide association studies (GWAS) have found a significant correlation between increased frequency of single-nucleotide polymorphisms (SNPs) in the Cdkn1a gene (encoding p21Cip1/Waf1) that leads to increased p21Cip1/Waf1 expression in NAFLD [86, 87]. Increases in expression of p21Cip1/Waf1 is one of the main markers of senescence [87], because it is a potent negative regulator of CDK1 activity. Moreover, our data demonstrates that long term deletion of Cdk1 in hepatic cells induced hepatic expression of p21Cip1/Waf1, cellular senescence, fibrosis and inflammation (unpublished data). In this section we will summarize the current role of hepatic cell cycle regulators in lipid biosynthesis, senescence and onset of NAFLD.

The p21-Cdk1-E2F1 axis

Recent findings have indicated that NAFLD livers display lower expression of Cdk1 [88]. CDK1 is essential for mitotic division, but in hepatic cells CDK1 may have other functions beyond mitosis. For example, we have shown that deletion of Cdk1 in hepatocytes leads to increases in nuclear and cell size [21]. DNA re-replication by endoreduplication may be the underlying cause of this effect. During endoreduplication, hepatic cells may generate oxidative stress leading to chronic liver damage [88]. In this setting, p21Cip1/Waf1 negatively regulates CDK1, and therefore increased levels of p21Cip1/Waf1 in liver disease may correlate with reduced activity of CDK1 [88]. Therefore, it may be therapeutically feasible to reduce the expression of p21Cip1/Waf1 in hepatocytes to reduce NAFLD. However, the pleiotropic functions of p21Cip1/Waf1 are likely to make it an “undruggable” target. Interestingly, it was recently shown that senotherapeutic drugs improved liver regeneration when p21 was overexpressed further suggesting that SASP may inhibit liver regeneration [89].
An important function for p21Cip1/Waf1 is the transcriptional regulation of cell cycle genes. Typically, p21Cip1/Waf1 binds with high affinity to and inhibits CDK2/cyclin E activity [39] which leads to hypo-phosphorylation of the RB protein [90]. Hypophosphorylated RB remains bound to E2F1, repressing its activation and cell cycle progression. Recent results highlighted a number of novel metabolic functions of E2F1 [91]. For example, E2F1 mRNA and protein are increased in NASH livers [92]. On top of that, hepatic deletion of E2f1 significantly decreased fibrosis as a consequence of downregulation of the Egr-1 gene [93, 94]. Moreover, E2F1 regulates the expression of essential genes in lipid synthesis such as Acaca, Fasn, Scd1, Srebp1c, and Chrebp [92]. The interaction of p21Cip1/Waf1 with CDK1 may lead to inactivation of E2F1. E2F1 also drives the expression of Cdk1 [94]. In this fashion, p21Cip1/Waf1 regulates cell cycle progression (Fig. 5), cellular division, senescence and metabolism through the expression of numerous genes, including lipogenic enzymes {for review see [95]}.
Finally, the transcription of p21Cip1/Waf1 is activated by p53. In response to DNA damage, p53 promotes the expression of p21Cip1/Waf1, leading to cell cycle arrest and senescence [22, 96, 97]. Simultaneously, p53 regulates the transcription of many genes involved in glycolysis, lipogenesis and other metabolic pathways [98]. Particularly interesting is the discovery of frequent SNPs variants in p53 that predispose patients to develop obesity and NAFLD [99] (Fig. 5). When these variants were introduced in mice fed with HFD, they caused significant increases in body weight with liver steatosis, inflammation, and fibrosis [100]. These mice also displayed a remarkable increase in the expression of p21Cip1/Waf1 and inflammatory genes. Moreover, under fasting conditions, hepatic cells accumulate lipid droplets which correlate with increased expression of both p53 [99, 100] and p21 [86, 87]. Collectively, these observations suggest that p53 may fulfill essential roles upstream of p21 Cip1/Waf1 and Cdk1 in NAFLD.

Conclusion and future perspective

The regulation of cell cycle progression, its components and the interactions among them has been meticulously studied for more than four decades. Most of the basic functions discovered in vitro have been validated in vivo with similar physiological significance. However, an increasing number of studies now indicate that a large number of cell cycle regulators, including CDK1, influence metabolism which are new interactions that may not depend on cell cycle progression. This may not be surprising as cells need to integrate their growth with cell proliferation, into a wider network of biosynthetic pathways. However, this point has been largely ignored as the focus has been in the upstream signaling pathways regulating cell cycle progression. Studying the interplay between cell cycle and metabolism in detail will lead to development of more refined therapies for the treatment of diseases.
Particularly important is to understand the role of key cell cycle regulators CDK1 and p21 in regulating metabolic pathways during acute and chronic liver damage. The reasoning behind this is that if there is a distinction between the pro-regenerative functions of these two, and other functions, then it may be able to target them in NAFLD. It is well established that liver regeneration is dramatically reduced in patients with NAFLD, and therefore resection and transplantation is not an ideal therapy for this group of patients. This is exacerbated in the more advanced stage of disease, like NASH. In this setting, targeting pro-regenerative pathways of hepatocytes may lead to a better outcome. Our group has actively studied the non-canonical functions of Cdk1, but those of p21 have been neglected. For example, we have preliminary data indicating that downregulation of Cdk1 may lead to a NAFLD-like phenotype in the long term, with fibrosis and steatosis. Thus, in NAFLD patients it would be therapeutically relevant to disrupt the p21-Cdk1 axis, with increasing Cdk1 functions while reducing those of p21. However, there is a gap on our understanding of the functions of increased expression of p21 and its activity in the context of deletion of Cdk1, or if the observed phenotype is a Cdk1-autonomous effect.
Finally, studies of NAFLD have focused mostly on targeting inflammatory or non- parenchymal cells. Thus, studying the cell cycle and non-canonical functions of p21 and Cdk1 in hepatocytes may lead to far reaching therapeutic approaches for either early NAFLD or specially advanced NASH. Ideally personalized therapies may be developed, in which expression of p21 would be reduced in patients with SNPs in the promoter region. Following this approach, we will be able to reduce the signaling associated with increased p21 activity. These future discoveries will have direct implications in the development of therapeutic agents targeting both, metabolism and cell division, for these common liver diseases, NAFLD and NASH.

Acknowledgements

Open access funding provided by Lund University. We thank all present and past members of the Kaldis laboratory for discussions, input, and support. We also thank the administrative staff from the Host Defense Laboratory and the Immunology Frontiers Research Center for their support.

Compliance with ethical standards

Conflict of interest

Matias J. Caldez, Mikael Bjorklund and Philipp Kaldis declare no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Tsung A, Geller DA. Gross and cellular anatomy of the liver. In: Monga SPS, editor. Molecular pathology of liver diseases. Pittsburg: Springer; 2011. p. 3–6. Tsung A, Geller DA. Gross and cellular anatomy of the liver. In: Monga SPS, editor. Molecular pathology of liver diseases. Pittsburg: Springer; 2011. p. 3–6.
2.
Zurück zum Zitat Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology 2006;43(2 Suppl 1):S45–S53PubMed Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology 2006;43(2 Suppl 1):S45–S53PubMed
3.
Zurück zum Zitat Valizadeh A, et al. The roles of signaling pathways in liver repair and regeneration. J Cell Physiol. 2019;234(9):14966–74. Valizadeh A, et al. The roles of signaling pathways in liver repair and regeneration. J Cell Physiol. 2019;234(9):14966–74.
4.
Zurück zum Zitat Forbes SJ, Newsome PN. Liver regeneration—mechanisms and models to clinical application. Nat Rev Gastroenterol Hepatol 2016;13(8):473–485PubMed Forbes SJ, Newsome PN. Liver regeneration—mechanisms and models to clinical application. Nat Rev Gastroenterol Hepatol 2016;13(8):473–485PubMed
5.
Zurück zum Zitat Brunt EM, et al. Nonalcoholic fatty liver disease. Nat Rev Dis Prim 2015;1:15080PubMed Brunt EM, et al. Nonalcoholic fatty liver disease. Nat Rev Dis Prim 2015;1:15080PubMed
6.
Zurück zum Zitat Bhushan B, Udayan A. Liver regeneration after acetaminophen hepatotoxicuty: mechanisms and therapeutic opportunities. Am J Pathol 2019;189(4):719–729PubMedPubMedCentral Bhushan B, Udayan A. Liver regeneration after acetaminophen hepatotoxicuty: mechanisms and therapeutic opportunities. Am J Pathol 2019;189(4):719–729PubMedPubMedCentral
7.
Zurück zum Zitat Kurinna S, Barton MC. Cascades of transcription regulation during liver regeneration. Int J Biochem Cell Biol 2011;43(2):189–197PubMed Kurinna S, Barton MC. Cascades of transcription regulation during liver regeneration. Int J Biochem Cell Biol 2011;43(2):189–197PubMed
8.
Zurück zum Zitat Lorenz L, et al. Mechanosensing by beta1 integrin induces angiocrine signals for liver growth and survival. Nature 2018;562(7725):128–132PubMed Lorenz L, et al. Mechanosensing by beta1 integrin induces angiocrine signals for liver growth and survival. Nature 2018;562(7725):128–132PubMed
9.
Zurück zum Zitat Aldeguer X, et al. Interleukin-6 from intrahepatic cells of bone marrow origin is required for normal murine liver regeneration. Hepatology 2002;35(1):40–48PubMed Aldeguer X, et al. Interleukin-6 from intrahepatic cells of bone marrow origin is required for normal murine liver regeneration. Hepatology 2002;35(1):40–48PubMed
10.
Zurück zum Zitat Matsui T, et al. STAT3 down-regulates the expression of cyclin D during liver development. J Biol Chem 2002;277(39):36167–36173PubMed Matsui T, et al. STAT3 down-regulates the expression of cyclin D during liver development. J Biol Chem 2002;277(39):36167–36173PubMed
11.
Zurück zum Zitat Albrecht J, et al. Regulation of G(1) cyclin-dependent kinases in the liver: role of nuclear localization and p27 sequestration. Am J Physiol 1999;277(6):G1207–G1216PubMed Albrecht J, et al. Regulation of G(1) cyclin-dependent kinases in the liver: role of nuclear localization and p27 sequestration. Am J Physiol 1999;277(6):G1207–G1216PubMed
12.
Zurück zum Zitat Albrecht J, et al. Involvement of p21 and p27 in the regulation of CDK activity and cell cycle progression in the regenerating liver. Oncogene 1998;16(16):2141–2150PubMed Albrecht J, et al. Involvement of p21 and p27 in the regulation of CDK activity and cell cycle progression in the regenerating liver. Oncogene 1998;16(16):2141–2150PubMed
13.
Zurück zum Zitat Albrecht J, Hu M, Cerra F. Distinct patterns of cyclin D1 regulation in models of liver regeneration and human liver. Biochem Biophys Res Commun 1995;209(2):648–655PubMed Albrecht J, Hu M, Cerra F. Distinct patterns of cyclin D1 regulation in models of liver regeneration and human liver. Biochem Biophys Res Commun 1995;209(2):648–655PubMed
14.
Zurück zum Zitat Factor VM, et al. Loss of c-Met disrupts gene expression program required for G2/M progression during liver regeneration in mice. PLoS One. 2010;5(9):e12739.PubMedPubMedCentral Factor VM, et al. Loss of c-Met disrupts gene expression program required for G2/M progression during liver regeneration in mice. PLoS One. 2010;5(9):e12739.PubMedPubMedCentral
15.
Zurück zum Zitat Borowiak M, et al. Met provides essential signals for liver regeneration. Proc Natl Acad Sci USA 2004;101(29):10608–10613PubMedPubMedCentral Borowiak M, et al. Met provides essential signals for liver regeneration. Proc Natl Acad Sci USA 2004;101(29):10608–10613PubMedPubMedCentral
16.
Zurück zum Zitat Chauhan A, et al. Regulation of mammalian cell cycle progression in the regenerating liver. J Theor Biol 2011;283(1):103–112PubMed Chauhan A, et al. Regulation of mammalian cell cycle progression in the regenerating liver. J Theor Biol 2011;283(1):103–112PubMed
17.
Zurück zum Zitat Arai M, et al. Gene expression profiling reveals the mechanism and pathophysiology of mouse liver regeneration. J Biol Chem 2003;278(32):29813–29818PubMed Arai M, et al. Gene expression profiling reveals the mechanism and pathophysiology of mouse liver regeneration. J Biol Chem 2003;278(32):29813–29818PubMed
18.
Zurück zum Zitat Hohmann N, et al. How does a single cell know when the liver has reached its correct size? PLoS ONE 2014;9(4):e93207PubMedPubMedCentral Hohmann N, et al. How does a single cell know when the liver has reached its correct size? PLoS ONE 2014;9(4):e93207PubMedPubMedCentral
19.
Zurück zum Zitat Miyaoka Y, et al. Hypertrophy and unconventional cell division of hepatocytes underlie liver regeneration. Curr Biol 2012;22(13):1166–1175PubMed Miyaoka Y, et al. Hypertrophy and unconventional cell division of hepatocytes underlie liver regeneration. Curr Biol 2012;22(13):1166–1175PubMed
20.
Zurück zum Zitat Hui T, et al. Immediate early genes and p21 regulation in liver of rats with acute hepatic failure. Am J Surg 2002;183(4):457–463PubMed Hui T, et al. Immediate early genes and p21 regulation in liver of rats with acute hepatic failure. Am J Surg 2002;183(4):457–463PubMed
21.
Zurück zum Zitat Diril MK, et al. Cyclin-dependent kinase 1 (Cdk1) is essential for cell division and suppression of DNA re-replication but not for liver regeneration. Proc Natl Acad Sci USA 2012;109(10):3826–3831PubMedPubMedCentral Diril MK, et al. Cyclin-dependent kinase 1 (Cdk1) is essential for cell division and suppression of DNA re-replication but not for liver regeneration. Proc Natl Acad Sci USA 2012;109(10):3826–3831PubMedPubMedCentral
22.
Zurück zum Zitat Kurinna S, et al. p53 regulates a mitotic transcription program and determines ploidy in normal mouse liver. Hepatology 2013;57(5):2004–2133PubMed Kurinna S, et al. p53 regulates a mitotic transcription program and determines ploidy in normal mouse liver. Hepatology 2013;57(5):2004–2133PubMed
23.
Zurück zum Zitat Kossatz U, et al. Skp2-dependent degradation of p27kip1 is essential for cell cycle progression. Genes Dev 2004;18(21):2602–2607PubMedPubMedCentral Kossatz U, et al. Skp2-dependent degradation of p27kip1 is essential for cell cycle progression. Genes Dev 2004;18(21):2602–2607PubMedPubMedCentral
24.
Zurück zum Zitat Haga S, et al. Compensatory recovery of liver mass by Akt-mediated hepatocellular hypertrophy in liver-specific STAT3-deficient mice. J Hepatol 2005;43(5):799–807PubMed Haga S, et al. Compensatory recovery of liver mass by Akt-mediated hepatocellular hypertrophy in liver-specific STAT3-deficient mice. J Hepatol 2005;43(5):799–807PubMed
25.
Zurück zum Zitat Haga S, et al. The survival pathways phosphatidylinositol-3 kinase (PI3-K)/phosphoinositide-dependent protein kinase 1 (PDK1)/Akt modulate liver regeneration through hepatocyte size rather than proliferation. Hepatology 2009;49(1):204–214PubMed Haga S, et al. The survival pathways phosphatidylinositol-3 kinase (PI3-K)/phosphoinositide-dependent protein kinase 1 (PDK1)/Akt modulate liver regeneration through hepatocyte size rather than proliferation. Hepatology 2009;49(1):204–214PubMed
26.
Zurück zum Zitat Caldez MJ, et al. Metabolic remodeling during liver regeneration. Dev Cell 2018;47(4):425–438 (e5) PubMed Caldez MJ, et al. Metabolic remodeling during liver regeneration. Dev Cell 2018;47(4):425–438 (e5) PubMed
27.
Zurück zum Zitat Hydbring P, Malumbres M, Sicinski P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol 2016;17(5):280–292PubMedPubMedCentral Hydbring P, Malumbres M, Sicinski P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol 2016;17(5):280–292PubMedPubMedCentral
28.
Zurück zum Zitat Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013;140(15):3079–3093PubMed Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013;140(15):3079–3093PubMed
29.
Zurück zum Zitat Ilyin G, et al. Regulation and role of p21 and p27 cyclin-dependent kinase inhibitors during hepatocyte differentiation and growth. Am J Gastrointest Liver Physiol 2003;285(1):G115–G117 Ilyin G, et al. Regulation and role of p21 and p27 cyclin-dependent kinase inhibitors during hepatocyte differentiation and growth. Am J Gastrointest Liver Physiol 2003;285(1):G115–G117
30.
Zurück zum Zitat Loyer P, et al. Expression and activation of cdks (1 and 2) and cyclins in the cell cycle progression during liver regeneration. J Biol Chem 1994;269(4):2491–2500PubMed Loyer P, et al. Expression and activation of cdks (1 and 2) and cyclins in the cell cycle progression during liver regeneration. J Biol Chem 1994;269(4):2491–2500PubMed
31.
Zurück zum Zitat Rohlfing AK, et al. Partial hepatectomy in rats results in immediate down-regulation of p27Kip1 in residual liver tissue by transcriptional and post-translational processes. Front Physiol 2013;4:139PubMedPubMedCentral Rohlfing AK, et al. Partial hepatectomy in rats results in immediate down-regulation of p27Kip1 in residual liver tissue by transcriptional and post-translational processes. Front Physiol 2013;4:139PubMedPubMedCentral
32.
Zurück zum Zitat Chauhan A, et al. A mesoscale model of G1/S phase transition in liver regeneration. J Theor Biol 2008;252(3):465–473PubMed Chauhan A, et al. A mesoscale model of G1/S phase transition in liver regeneration. J Theor Biol 2008;252(3):465–473PubMed
33.
Zurück zum Zitat Pujol MJ, et al. Differential association of p21Cip1 and p27Kip1 with cyclin E-CDK2 during rat liver regeneration. J Hepatol 2000;33(3):266–274PubMed Pujol MJ, et al. Differential association of p21Cip1 and p27Kip1 with cyclin E-CDK2 during rat liver regeneration. J Hepatol 2000;33(3):266–274PubMed
34.
Zurück zum Zitat Trembley JH, Kren BT, Steer CJ. Posttranscriptional regulation of cyclin B messenger RNA expression in the regenerating rat liver. Cell Growth Differ 1994;5(1):99–108PubMed Trembley JH, Kren BT, Steer CJ. Posttranscriptional regulation of cyclin B messenger RNA expression in the regenerating rat liver. Cell Growth Differ 1994;5(1):99–108PubMed
35.
Zurück zum Zitat Trembley JH, et al. Differential regulation of cyclin B1 RNA and protein expression during hepatocyte growth in vivo. Cell Growth Differ 1996;7(7):903–916PubMed Trembley JH, et al. Differential regulation of cyclin B1 RNA and protein expression during hepatocyte growth in vivo. Cell Growth Differ 1996;7(7):903–916PubMed
36.
Zurück zum Zitat Spiewak Rinaudo JA, Thorgeirsson SS. Detection of a tyrosine-phosphorylated form of cyclin A during liver regeneration. Cell Growth Differ 1997;8(3):301–309PubMed Spiewak Rinaudo JA, Thorgeirsson SS. Detection of a tyrosine-phosphorylated form of cyclin A during liver regeneration. Cell Growth Differ 1997;8(3):301–309PubMed
37.
Zurück zum Zitat Verges M, et al. Cyclin A is present in the endocytic compartment of rat liver cells and increases during liver regeneration. Biochem Biophys Res Commun 1997;230(1):49–53PubMed Verges M, et al. Cyclin A is present in the endocytic compartment of rat liver cells and increases during liver regeneration. Biochem Biophys Res Commun 1997;230(1):49–53PubMed
38.
Zurück zum Zitat Garnier D, et al. Cyclin-dependent kinase 1 plays a critical role in DNA replication control during rat liver regeneration. Hepatology 2009;50(6):1946–1956PubMed Garnier D, et al. Cyclin-dependent kinase 1 plays a critical role in DNA replication control during rat liver regeneration. Hepatology 2009;50(6):1946–1956PubMed
39.
Zurück zum Zitat Satyanarayana A, Hilton MB, Kaldis P. p21 Inhibits Cdk1 in the absence of Cdk2 to maintain the G1/S phase DNA damage checkpoint. Mol Biol Cell 2008;19(1):65–77PubMedPubMedCentral Satyanarayana A, Hilton MB, Kaldis P. p21 Inhibits Cdk1 in the absence of Cdk2 to maintain the G1/S phase DNA damage checkpoint. Mol Biol Cell 2008;19(1):65–77PubMedPubMedCentral
40.
Zurück zum Zitat Szmyd R, et al. Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality. Oncogene 2019;38(7):998–1018PubMed Szmyd R, et al. Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality. Oncogene 2019;38(7):998–1018PubMed
42.
Zurück zum Zitat Moncada S, Higgs EA, Colombo SL. Fulfilling the metabolic requirements for cell proliferation. Biochem J 2012;446(1):1–7PubMed Moncada S, Higgs EA, Colombo SL. Fulfilling the metabolic requirements for cell proliferation. Biochem J 2012;446(1):1–7PubMed
43.
Zurück zum Zitat Anderson SP, et al. Delayed liver regeneration in peroxisome proliferator-activated receptor-alpha-null mice. Hepatology 2002;36(3):544–554PubMed Anderson SP, et al. Delayed liver regeneration in peroxisome proliferator-activated receptor-alpha-null mice. Hepatology 2002;36(3):544–554PubMed
44.
Zurück zum Zitat Espeillac C, et al. S6 kinase 1 is required for rapamycin-sensitive liver proliferation after mouse hepatectomy. J Clin Investig 2011;121(7):2821–2832PubMedPubMedCentral Espeillac C, et al. S6 kinase 1 is required for rapamycin-sensitive liver proliferation after mouse hepatectomy. J Clin Investig 2011;121(7):2821–2832PubMedPubMedCentral
45.
Zurück zum Zitat Huang W, et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 2006;312(5771):233–236PubMed Huang W, et al. Nuclear receptor-dependent bile acid signaling is required for normal liver regeneration. Science 2006;312(5771):233–236PubMed
46.
Zurück zum Zitat Varela-Rey M, et al. Impaired liver regeneration in mice lacking glycine N-methyltransferase. Hepatology 2009;50(2):443–452PubMed Varela-Rey M, et al. Impaired liver regeneration in mice lacking glycine N-methyltransferase. Hepatology 2009;50(2):443–452PubMed
47.
Zurück zum Zitat Mukherjee S, et al. Nicotinamide adenine dinucleotide biosynthesis promotes liver regeneration. Hepatology 2017;65(2):616–630PubMed Mukherjee S, et al. Nicotinamide adenine dinucleotide biosynthesis promotes liver regeneration. Hepatology 2017;65(2):616–630PubMed
48.
Zurück zum Zitat Meng Z, et al. Insufficient bile acid signaling impairs liver repair in CYP27(−/−) mice. J Hepatol 2011;55(4):885–895PubMed Meng Z, et al. Insufficient bile acid signaling impairs liver repair in CYP27(−/−) mice. J Hepatol 2011;55(4):885–895PubMed
49.
Zurück zum Zitat Crumm S, et al. Adenine nucleotide changes in the remnant liver: an early signal for regeneration after partial hepatectomy. Hepatology 2008;48(3):898–908PubMed Crumm S, et al. Adenine nucleotide changes in the remnant liver: an early signal for regeneration after partial hepatectomy. Hepatology 2008;48(3):898–908PubMed
50.
Zurück zum Zitat Kachaylo E, et al. PTEN down-regulation promotes beta-oxidation to fuel hypertrophic liver growth after hepatectomy in mice. Hepatology 2017;66(3):908–921PubMed Kachaylo E, et al. PTEN down-regulation promotes beta-oxidation to fuel hypertrophic liver growth after hepatectomy in mice. Hepatology 2017;66(3):908–921PubMed
51.
Zurück zum Zitat Fernandez M, et al. Caveolin-1 is essential for liver regeneration. Science 2006;313(5793):1628–1632PubMed Fernandez M, et al. Caveolin-1 is essential for liver regeneration. Science 2006;313(5793):1628–1632PubMed
52.
Zurück zum Zitat Shteyer E, et al. Disruption of hepatic adipogenesis is associated with impaired liver regeneration in mice. Hepatology 2004;40(6):1322–1332PubMed Shteyer E, et al. Disruption of hepatic adipogenesis is associated with impaired liver regeneration in mice. Hepatology 2004;40(6):1322–1332PubMed
53.
Zurück zum Zitat Garcia-Arcos I, et al. Lipid analysis reveals quiescent and regenerating liver-specific populations of lipid droplets. Lipids 2010;45(12):1101–1108PubMed Garcia-Arcos I, et al. Lipid analysis reveals quiescent and regenerating liver-specific populations of lipid droplets. Lipids 2010;45(12):1101–1108PubMed
54.
Zurück zum Zitat Huang J, et al. Postponing the hypoglycemic response to partial hepatectomy delays mouse liver regeneration. Am J Pathol 2016;186(3):587–599PubMedPubMedCentral Huang J, et al. Postponing the hypoglycemic response to partial hepatectomy delays mouse liver regeneration. Am J Pathol 2016;186(3):587–599PubMedPubMedCentral
55.
Zurück zum Zitat Wang Z, et al. Cyclin B1/Cdk1 coordinates mitochondrial respiration for cell-cycle G2/M progression. Dev Cell 2014;29(2):217–232PubMedPubMedCentral Wang Z, et al. Cyclin B1/Cdk1 coordinates mitochondrial respiration for cell-cycle G2/M progression. Dev Cell 2014;29(2):217–232PubMedPubMedCentral
56.
Zurück zum Zitat Qin L, et al. CDK1 enhances mitochondrial bioenergetics for radiation-induced DNA repair. Cell Rep 2015;13(10):2056–2063PubMedPubMedCentral Qin L, et al. CDK1 enhances mitochondrial bioenergetics for radiation-induced DNA repair. Cell Rep 2015;13(10):2056–2063PubMedPubMedCentral
57.
Zurück zum Zitat Yuan Z, et al. Activation of FOXO1 by Cdk1 in cycling cells and postmitotic neurons. Science 2008;319(5870):1665–1668PubMed Yuan Z, et al. Activation of FOXO1 by Cdk1 in cycling cells and postmitotic neurons. Science 2008;319(5870):1665–1668PubMed
58.
Zurück zum Zitat Yalcin A, et al. 6-Phosphofructo-2-kinase (PFKFB3) promotes cell cycle progression and suppresses apoptosis via Cdk1-mediated phosphorylation of p27. Cell Death Dis 2014;5:e1337PubMedPubMedCentral Yalcin A, et al. 6-Phosphofructo-2-kinase (PFKFB3) promotes cell cycle progression and suppresses apoptosis via Cdk1-mediated phosphorylation of p27. Cell Death Dis 2014;5:e1337PubMedPubMedCentral
59.
Zurück zum Zitat Yalcin A, et al. Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases. J Biol Chem 2009;284(36):24223–24232PubMedPubMedCentral Yalcin A, et al. Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases. J Biol Chem 2009;284(36):24223–24232PubMedPubMedCentral
60.
Zurück zum Zitat Miettinen TP, et al. Identification of transcriptional and metabolic programs related to mammalian cell size. Curr Biol 2014;24(6):598–608PubMedPubMedCentral Miettinen TP, et al. Identification of transcriptional and metabolic programs related to mammalian cell size. Curr Biol 2014;24(6):598–608PubMedPubMedCentral
61.
Zurück zum Zitat Tu BP, et al. Cyclic changes in metabolic state during the life of a yeast cell. Proc Natl Acad Sci USA 2007;104(43):16886–16891PubMedPubMedCentral Tu BP, et al. Cyclic changes in metabolic state during the life of a yeast cell. Proc Natl Acad Sci USA 2007;104(43):16886–16891PubMedPubMedCentral
62.
Zurück zum Zitat Chen Z, et al. Restriction of DNA replication to the reductive phase of the metabolic cycle protects genome integrity. Science 2007;316(5833):1916–1919PubMed Chen Z, et al. Restriction of DNA replication to the reductive phase of the metabolic cycle protects genome integrity. Science 2007;316(5833):1916–1919PubMed
63.
Zurück zum Zitat Huang J, Borensztajn J, Reddy JK. Hepatic lipid metabolism. In: Monga SPS, editor. Molecular pathology of liver diseases. Pittsburg: Springer; 2011. p. 133–146 Huang J, Borensztajn J, Reddy JK. Hepatic lipid metabolism. In: Monga SPS, editor. Molecular pathology of liver diseases. Pittsburg: Springer; 2011. p. 133–146
64.
Zurück zum Zitat Hijmans BS, et al. Zonation of glucose and fatty acid metabolism in the liver: mechanism and metabolic consequences. Biochimie 2014;96:121–129PubMed Hijmans BS, et al. Zonation of glucose and fatty acid metabolism in the liver: mechanism and metabolic consequences. Biochimie 2014;96:121–129PubMed
65.
Zurück zum Zitat Peng J, et al. Enhanced liver regeneration after partial hepatectomy in sterol regulatory element-binding protein (SREBP)-1c-null mice is associated with increased hepatocellular cholesterol availability. Cell Physiol Biochem 2018;47(2):784–799PubMed Peng J, et al. Enhanced liver regeneration after partial hepatectomy in sterol regulatory element-binding protein (SREBP)-1c-null mice is associated with increased hepatocellular cholesterol availability. Cell Physiol Biochem 2018;47(2):784–799PubMed
66.
Zurück zum Zitat Martínez-Botas J, et al. Cholesterol starvation decreases p34(cdc2) kinase activity and arrests the cell cycle at G2. FASEB J. 1999;13(11):1359–70.PubMed Martínez-Botas J, et al. Cholesterol starvation decreases p34(cdc2) kinase activity and arrests the cell cycle at G2. FASEB J. 1999;13(11):1359–70.PubMed
67.
Zurück zum Zitat Fernandez C, et al. Cholesterol is essential for mitosis progression and its deficiency induces polyploid cell formation. Exp Cell Res 2004;300(1):109–120PubMed Fernandez C, et al. Cholesterol is essential for mitosis progression and its deficiency induces polyploid cell formation. Exp Cell Res 2004;300(1):109–120PubMed
68.
Zurück zum Zitat Atilla-Gokcumen GE, et al. Dividing cells regulate their lipid composition and localization. Cell 2014;156(3):428–439PubMedPubMedCentral Atilla-Gokcumen GE, et al. Dividing cells regulate their lipid composition and localization. Cell 2014;156(3):428–439PubMedPubMedCentral
69.
Zurück zum Zitat McCormack L, et al. Hepatic steatosis is a risk factor for postoperative complications after major hepatectomy: a matched case-control study. Ann Surg 2007;245(6):923–930PubMedPubMedCentral McCormack L, et al. Hepatic steatosis is a risk factor for postoperative complications after major hepatectomy: a matched case-control study. Ann Surg 2007;245(6):923–930PubMedPubMedCentral
70.
Zurück zum Zitat Dutkowski P, et al. Challenges to liver transplantation and strategies to improve outcomes. Gastroenterology 2015;148(2):307–323PubMed Dutkowski P, et al. Challenges to liver transplantation and strategies to improve outcomes. Gastroenterology 2015;148(2):307–323PubMed
71.
Zurück zum Zitat Vetelainen R, van Vliet AK, van Gulik TM. Severe steatosis increases hepatocellular injury and impairs liver regeneration in a rat model of partial hepatectomy. Ann Surg 2007;245(1):44–50PubMedPubMedCentral Vetelainen R, van Vliet AK, van Gulik TM. Severe steatosis increases hepatocellular injury and impairs liver regeneration in a rat model of partial hepatectomy. Ann Surg 2007;245(1):44–50PubMedPubMedCentral
72.
Zurück zum Zitat Mendez-Sanchez N, et al. New aspects of lipotoxicity in nonalcoholic steatohepatitis. Int J Mol Sci. 2018;19(7):2034.PubMedCentral Mendez-Sanchez N, et al. New aspects of lipotoxicity in nonalcoholic steatohepatitis. Int J Mol Sci. 2018;19(7):2034.PubMedCentral
73.
Zurück zum Zitat Perry RJ, et al. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation. Cell 2018;172(1–2):234–248PubMedPubMedCentral Perry RJ, et al. Leptin mediates a glucose-fatty acid cycle to maintain glucose homeostasis in starvation. Cell 2018;172(1–2):234–248PubMedPubMedCentral
74.
Zurück zum Zitat Selzner M, Clavein P. Failure of regeneration of the steatotic rat liver: disruption at two different levels in the regeneration pathway. Hepatology 2000;31(1):35–42PubMed Selzner M, Clavein P. Failure of regeneration of the steatotic rat liver: disruption at two different levels in the regeneration pathway. Hepatology 2000;31(1):35–42PubMed
75.
Zurück zum Zitat Yang SQ, et al. Disrupted signaling and inhibited regeneration in obese mice with fatty livers: implications for nonalcoholic fatty liver disease pathophysiology. Hepatology 2001;34(4 Pt 1):694–706PubMed Yang SQ, et al. Disrupted signaling and inhibited regeneration in obese mice with fatty livers: implications for nonalcoholic fatty liver disease pathophysiology. Hepatology 2001;34(4 Pt 1):694–706PubMed
76.
Zurück zum Zitat Pauta M, et al. Impaired liver regeneration in Ldlr−/− mice is associated with an altered hepatic profile of cytokines, growth factors, and lipids. J Hepatol 2013;59(4):731–737PubMedPubMedCentral Pauta M, et al. Impaired liver regeneration in Ldlr−/− mice is associated with an altered hepatic profile of cytokines, growth factors, and lipids. J Hepatol 2013;59(4):731–737PubMedPubMedCentral
77.
Zurück zum Zitat Leclercq IA, Field J, Farrell GC. Leptin-specific mechanisms for impaired liver regeneration in ob/ob mice after toxic injury. Gastroenterology 2003;124(5):1451–1464PubMed Leclercq IA, Field J, Farrell GC. Leptin-specific mechanisms for impaired liver regeneration in ob/ob mice after toxic injury. Gastroenterology 2003;124(5):1451–1464PubMed
78.
Zurück zum Zitat Leclercq IA, et al. Defective hepatic regeneration after partial hepatectomy in leptin-deficient mice is not rescued by exogenous leptin. Lab Investig 2006;86(11):1161–1171PubMed Leclercq IA, et al. Defective hepatic regeneration after partial hepatectomy in leptin-deficient mice is not rescued by exogenous leptin. Lab Investig 2006;86(11):1161–1171PubMed
79.
Zurück zum Zitat Picard C, et al. Steatosis is not sufficient to cause an impaired regenerative response after partial hepatectomy in rats. J Hepatol 2002;36(5):645–652PubMed Picard C, et al. Steatosis is not sufficient to cause an impaired regenerative response after partial hepatectomy in rats. J Hepatol 2002;36(5):645–652PubMed
80.
Zurück zum Zitat Zhao J, et al. NAFLD induction delays postoperative liver regeneration of ALPPS in rats. Dig Dis Sci 2019;64(2):456–468PubMed Zhao J, et al. NAFLD induction delays postoperative liver regeneration of ALPPS in rats. Dig Dis Sci 2019;64(2):456–468PubMed
81.
Zurück zum Zitat Murata H, et al. Mechanism of impaired regeneration of fatty liver in mouse partial hepatectomy model. J Gastroenterol Hepatol 2007;22(12):2173–2180PubMed Murata H, et al. Mechanism of impaired regeneration of fatty liver in mouse partial hepatectomy model. J Gastroenterol Hepatol 2007;22(12):2173–2180PubMed
82.
Zurück zum Zitat DeAngelis RA, et al. A high-fat diet impairs liver regeneration in C57BL/6 mice through overexpression of the NF-kappaB inhibitor IkappaBalpha. Hepatology 2005;42(5):1148–1157PubMed DeAngelis RA, et al. A high-fat diet impairs liver regeneration in C57BL/6 mice through overexpression of the NF-kappaB inhibitor IkappaBalpha. Hepatology 2005;42(5):1148–1157PubMed
83.
Zurück zum Zitat Newberry EP, et al. Altered hepatic triglyceride content after partial hepatectomy without impaired liver regeneration in multiple murine genetic models. Hepatology 2008;48(4):1097–1105PubMed Newberry EP, et al. Altered hepatic triglyceride content after partial hepatectomy without impaired liver regeneration in multiple murine genetic models. Hepatology 2008;48(4):1097–1105PubMed
84.
Zurück zum Zitat Hamano M, et al. Lipid overloading during liver regeneration causes delayed hepatocyte DNA replication by increasing ER stress in mice with simple hepatic steatosis. J Gastroenterol 2014;49(2):305–316PubMed Hamano M, et al. Lipid overloading during liver regeneration causes delayed hepatocyte DNA replication by increasing ER stress in mice with simple hepatic steatosis. J Gastroenterol 2014;49(2):305–316PubMed
85.
Zurück zum Zitat Gazit V, et al. Liver regeneration is impaired in lipodystrophic fatty liver dystrophy mice. Hepatology 2010;52(6):2109–2117PubMed Gazit V, et al. Liver regeneration is impaired in lipodystrophic fatty liver dystrophy mice. Hepatology 2010;52(6):2109–2117PubMed
86.
Zurück zum Zitat Aravinthan A, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol 2013;58(3):549–556PubMed Aravinthan A, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol 2013;58(3):549–556PubMed
87.
Zurück zum Zitat Aravinthan A, et al. Gene polymorphisms of cellular senescence marker p21 and disease progression in non-alcohol-related fatty liver disease. Cell Cycle 2014;13(9):1489–1494PubMedPubMedCentral Aravinthan A, et al. Gene polymorphisms of cellular senescence marker p21 and disease progression in non-alcohol-related fatty liver disease. Cell Cycle 2014;13(9):1489–1494PubMedPubMedCentral
88.
Zurück zum Zitat Gentric G, et al. Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease. J Clin Investig 2015;125(3):981–992PubMedPubMedCentral Gentric G, et al. Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease. J Clin Investig 2015;125(3):981–992PubMedPubMedCentral
89.
Zurück zum Zitat Ritschka B, et al. The senotherapeutic drug ABT-737 disrupts aberrant p21 expression to restore liver regeneration in adult mice. Genes Dev 2020;34(7–8):489–494PubMedPubMedCentral Ritschka B, et al. The senotherapeutic drug ABT-737 disrupts aberrant p21 expression to restore liver regeneration in adult mice. Genes Dev 2020;34(7–8):489–494PubMedPubMedCentral
90.
Zurück zum Zitat Rivadeneira DB, et al. Proliferative suppression by CDK4/6 inhibition: complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology 2010;138(5):1920–1930PubMed Rivadeneira DB, et al. Proliferative suppression by CDK4/6 inhibition: complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology 2010;138(5):1920–1930PubMed
91.
Zurück zum Zitat Conner EA, et al. Dual functions of E2F–1 in a transgenic mouse model of liver carcinogenesis. Oncogene 2000;19(44):5054–5062PubMed Conner EA, et al. Dual functions of E2F–1 in a transgenic mouse model of liver carcinogenesis. Oncogene 2000;19(44):5054–5062PubMed
92.
Zurück zum Zitat Denechaud PD, et al. E2F1 mediates sustained lipogenesis and contributes to hepatic steatosis. J Clin Investig 2016;126(1):137–150PubMed Denechaud PD, et al. E2F1 mediates sustained lipogenesis and contributes to hepatic steatosis. J Clin Investig 2016;126(1):137–150PubMed
93.
Zurück zum Zitat Liao Y, et al. Delayed hepatocellular mitotic progression and impaired liver regeneration in early growth response-1-deficient mice. J Biol Chem 2004;279(41):43107–43116PubMed Liao Y, et al. Delayed hepatocellular mitotic progression and impaired liver regeneration in early growth response-1-deficient mice. J Biol Chem 2004;279(41):43107–43116PubMed
94.
Zurück zum Zitat Zellmer S, et al. Transcription factors ETF, E2F, and SP-1 are involved in cytokine-independent proliferation of murine hepatocytes. Hepatology 2010;52(6):2127–2136PubMed Zellmer S, et al. Transcription factors ETF, E2F, and SP-1 are involved in cytokine-independent proliferation of murine hepatocytes. Hepatology 2010;52(6):2127–2136PubMed
96.
Zurück zum Zitat Inoue Y, et al. Induction of p53-dependent p21 limits proliferative activity of rat hepatocytes in the presence of hepatocyte growth factor. PLoS ONE 2013;8(11):e78346PubMedPubMedCentral Inoue Y, et al. Induction of p53-dependent p21 limits proliferative activity of rat hepatocytes in the presence of hepatocyte growth factor. PLoS ONE 2013;8(11):e78346PubMedPubMedCentral
97.
Zurück zum Zitat Inoue Y, et al. p53 May positively regulate hepatocyte proliferation in rats. Hepatology 2002;36(2):336–344PubMed Inoue Y, et al. p53 May positively regulate hepatocyte proliferation in rats. Hepatology 2002;36(2):336–344PubMed
98.
Zurück zum Zitat Prokesch A, et al. Liver p53 is stabilized upon starvation and required for amino acid catabolism and gluconeogenesis. FASEB J 2017;31(2):732–742PubMed Prokesch A, et al. Liver p53 is stabilized upon starvation and required for amino acid catabolism and gluconeogenesis. FASEB J 2017;31(2):732–742PubMed
99.
Zurück zum Zitat Kung CP, et al. The P72R polymorphism of p53 predisposes to obesity and metabolic dysfunction. Cell Rep 2016;14(10):2413–2425PubMedPubMedCentral Kung CP, et al. The P72R polymorphism of p53 predisposes to obesity and metabolic dysfunction. Cell Rep 2016;14(10):2413–2425PubMedPubMedCentral
100.
Zurück zum Zitat Tomita K, et al. p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice. J Hepatol 2012;57(4):837–843PubMed Tomita K, et al. p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice. J Hepatol 2012;57(4):837–843PubMed
Metadaten
Titel
Cell cycle regulation in NAFLD: when imbalanced metabolism limits cell division
verfasst von
Matias J. Caldez
Mikael Bjorklund
Philipp Kaldis
Publikationsdatum
22.06.2020
Verlag
Springer India
Erschienen in
Hepatology International / Ausgabe 4/2020
Print ISSN: 1936-0533
Elektronische ISSN: 1936-0541
DOI
https://doi.org/10.1007/s12072-020-10066-6

Weitere Artikel der Ausgabe 4/2020

Hepatology International 4/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Aquatherapie bei Fibromyalgie wirksamer als Trockenübungen

03.05.2024 Fibromyalgiesyndrom Nachrichten

Bewegungs-, Dehnungs- und Entspannungsübungen im Wasser lindern die Beschwerden von Patientinnen mit Fibromyalgie besser als das Üben auf trockenem Land. Das geht aus einer spanisch-brasilianischen Vergleichsstudie hervor.

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.