Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.723
Filter
1.
Cell Death Dis ; 15(9): 681, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289348

ABSTRACT

Liver regeneration is an intricate pathophysiological process that has been a subject of great interest to the scientific community for many years. The capacity of liver regeneration is very critical for patients with liver diseases. Therefore, exploring the mechanisms of liver regeneration and finding good ways to improve it are very meaningful. Mesencephalic astrocyte-derived neurotrophic factor (MANF), a member of newly identified neurotrophic factors (NTFs) family, extensively expresses in the liver and has demonstrated cytoprotective effects during ER stress and inflammation. However, the role of MANF in liver regeneration remains unclear. Here, we used hepatocyte-specific MANF knockout (MANFHep-/-) mice to investigate the role of MANF in liver regeneration after 2/3 partial hepatectomy (PH). Our results showed that MANF expression was up-regulated in a time-dependent manner, and the peak level of mRNA and protein appeared at 24 h and 36 h after 2/3 PH, respectively. Notably, MANF knockout delayed hepatocyte proliferation, and the peak proliferation period was delayed by 24 h. Mechanistically, our in vitro results showed that MANF physically interacts with LRP5 and ß-catenin, two essential components of Wnt/ß-catenin pathway. Specifically, as a cofactor, MANF binds to the extracellular segment of LRP5 to activate Wnt/ß-catenin signaling. On the other hand, MANF interacts with ß-catenin to stabilize cytosolic ß-catenin level and promote its nuclear translocation, which further enhance the Wnt/ß-catenin signaling. We also found that MANF knockout does not affect the c-Met/ß-catenin complex after 2/3 PH. In summary, our study confirms that MANF may serve as a novel hepatocyte factor that is closely linked to the activation of the Wnt/ß-catenin pathway via intracellular and extracellular targets.


Subject(s)
Cell Proliferation , Hepatectomy , Hepatocytes , Liver Regeneration , Mice, Knockout , Nerve Growth Factors , Wnt Signaling Pathway , beta Catenin , Liver Regeneration/physiology , Animals , Nerve Growth Factors/metabolism , Nerve Growth Factors/genetics , Hepatocytes/metabolism , beta Catenin/metabolism , Mice , Humans , Mice, Inbred C57BL , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Male , Liver/metabolism
2.
PLoS One ; 19(9): e0310219, 2024.
Article in English | MEDLINE | ID: mdl-39259742

ABSTRACT

Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPß. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.


Subject(s)
Aging , GTP-Binding Proteins , Liver Regeneration , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc , Animals , Liver Regeneration/genetics , Liver Regeneration/physiology , Mice , Aging/metabolism , Aging/physiology , Aging/genetics , Humans , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Hepatectomy , Binding Sites , Liver/metabolism , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation , Transcription, Genetic , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Male , Carrier Proteins/metabolism , Carrier Proteins/genetics , Mice, Inbred C57BL , Cell Line, Tumor , RNA-Binding Proteins
3.
FASEB J ; 38(17): e70039, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39258958

ABSTRACT

Platelets play a crucial role in tissue regeneration, and their involvement in liver regeneration is well-established. However, the specific contribution of platelet-derived Transforming Growth Factor Beta 1 (TGFß1) to liver regeneration remains unexplored. This study investigated the role of platelet-derived TGFß1 in initiating liver regeneration following 2/3 liver resection. Using platelet-specific TGFß1 knockout (Plt.TGFß1 KO) mice and wild-type littermates (Plt.TGFß1 WT) as controls, the study assessed circulating levels and hepatic gene expression of TGFß1, Platelet Factor 4 (PF4), and Thrombopoietin (TPO) at early time points post-hepatectomy (post-PHx). Hepatocyte proliferation was quantified through Ki67 staining and PCNA expression in total liver lysates at various intervals, and phosphohistone-H3 (PHH3) staining was employed to mark mitotic cells. Circulating levels of hepatic mitogens, Hepatocyte Growth Factor (HGF), and Interleukin-6 (IL6) were also assessed. Results revealed that platelet-TGFß1 deficiency significantly reduced total plasma TGFß1 levels at 5 h post-PHx in Plt.TGFß1 KO mice compared to controls. While circulating PF4 levels, liver platelet recruitment and activation appeared normal at early time points, Plt.TGFß1 KO mice showed more stable circulating platelet numbers with higher numbers at 48 h post-PHx. Notably, hepatocyte proliferation was significantly reduced in Plt.TGFß1 KO mice. The results show that a lack of TGFß1 in platelets leads to an unbalanced expression of IL6 in the liver and to strongly increased HGF levels 48 h after liver resection, and yet liver regeneration remains reduced. The study identifies platelet-TGFß1 as a regulator of hepatocyte proliferation and platelet homeostasis in the early stages of liver regeneration.


Subject(s)
Blood Platelets , Hepatectomy , Liver Regeneration , Mice, Knockout , Thrombopoietin , Transforming Growth Factor beta1 , Animals , Liver Regeneration/physiology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Mice , Blood Platelets/metabolism , Thrombopoietin/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Cell Proliferation , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Liver/metabolism , Hepatocytes/metabolism , Male , Platelet Factor 4/metabolism , Platelet Factor 4/genetics , Mice, Inbred C57BL
4.
Int J Nanomedicine ; 19: 8117-8137, 2024.
Article in English | MEDLINE | ID: mdl-39139504

ABSTRACT

Background: The liver's regenerative capacity allows it to repair itself after injury. Extracellular vesicles and particles (EVPs) in the liver's interstitial space are crucial for signal transduction, metabolism, and immune regulation. Understanding the role and mechanism of liver-derived EVPs in regeneration is significant, particularly after partial hepatectomy, where the mechanisms remain unclear. Methods: A 70% hepatectomy model was established in mice, and EVPs were isolated and characterized using electron microscopy, nanocharacterization, and Western blot analysis. Combined metabolomic and transcriptomic analyses revealed ß-sitosterol enrichment in EVPs and activation of the Hedgehog signaling pathway during regeneration. The role of ß-sitosterol in EVPs on the Hedgehog pathway and its targets were identified using qRT-PCR, Western blot analysis. The regulation of carnitine synthesis by this pathway was determined using a dual luciferase assay. The effect of a ß-sitosterol diet on liver regeneration was verified in mice. Results: After 70% hepatectomy, the liver successfully regenerated without liver failure or death. At 24 hours post-surgery, tissue staining showed transient regeneration-associated steatosis (TRAS), with increased Ki67 positivity at 48 hours. EVPs displayed a spherical lipid bilayer structure with particle sizes of 70-130 nm. CD9, CD63, and CD81 in liver-derived EVPs were confirmed. Transcriptomic and metabolomic analyses showed EVPs supplementation significantly promoted carnitine synthesis and fatty acid oxidation. Tissue staining confirmed accelerated TRAS resolution and enhanced liver regeneration with EVP supplementation. Mass spectrometry identified ß-sitosterol in EVPs, which binds to Smo protein, activating the Hedgehog pathway. This led to the nuclear transport of Gli3, stimulating Setd5 transcription and inducing carnitine synthesis, thereby accelerating fatty acid oxidation. Mice with increased ß-sitosterol intake showed faster TRAS resolution and liver regeneration compared to controls. Conclusion: Liver-derived EVPs promote regeneration after partial hepatectomy. ß-sitosterol from EVPs accelerates fatty acid oxidation and promotes liver regeneration by activating Hedgehog signaling pathway.


Subject(s)
Extracellular Vesicles , Hedgehog Proteins , Hepatectomy , Liver Regeneration , Liver , Sitosterols , Animals , Sitosterols/pharmacology , Sitosterols/chemistry , Liver Regeneration/drug effects , Liver Regeneration/physiology , Extracellular Vesicles/drug effects , Extracellular Vesicles/chemistry , Mice , Liver/drug effects , Liver/metabolism , Hedgehog Proteins/metabolism , Male , Signal Transduction/drug effects , Mice, Inbred C57BL , Carnitine/pharmacology , Particle Size
5.
Clin Transl Med ; 14(8): e1812, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39152680

ABSTRACT

The liver possesses a distinctive capacity for regeneration within the human body. Under normal circumstances, liver cells replicate themselves to maintain liver function. Compensatory replication of healthy hepatocytes is sufficient for the regeneration after acute liver injuries. In the late stage of chronic liver damage, a large number of hepatocytes die and hepatocyte replication is blocked. Liver regeneration has more complex mechanisms, such as the transdifferentiation between cell types or hepatic progenitor cells mediated. Dysregulation of liver regeneration causes severe chronic liver disease. Gaining a more comprehensive understanding of liver regeneration mechanisms would facilitate the advancement of efficient therapeutic approaches. This review provides an overview of the signalling pathways linked to different aspects of liver regeneration in various liver diseases. Moreover, new knowledge on cellular interactions during the regenerative process is also presented. Finally, this paper explores the potential applications of new technologies, such as nanotechnology, stem cell transplantation and organoids, in liver regeneration after injury, offering fresh perspectives on treating liver disease.


Subject(s)
Liver Regeneration , Liver Regeneration/physiology , Humans , Liver Diseases/therapy , Liver Diseases/physiopathology , Cell Communication/physiology , Liver/injuries , Hepatocytes/metabolism , Signal Transduction , Animals
6.
Front Endocrinol (Lausanne) ; 15: 1404318, 2024.
Article in English | MEDLINE | ID: mdl-39145310

ABSTRACT

Background: Hepatocyte nuclear factor 4 alpha (HNF4α) is the master regulator of hepatic differentiation. Recent studies have also revealed the role of HNF4α in hepatocyte proliferation via negatively regulating the expression of proto-mitogenic genes, including cMyc. Here, we aimed to study the interaction between HNF4α-cMyc during liver regeneration after partial hepatectomy (PHX). Methods: Wild-type (WT), hepatocyte-specific knockout of HNF4α (HNF4α-KO), cMyc (cMyc-KO), and HNF4α-cMyc double knockout (DKO) mice were subjected to PHX to induce liver regeneration. Blood and liver tissue samples were collected at 0h, 24h, 48h, 7D, and 14D after PHX for further analysis. Results: WT, HNF4α-KO, cMyc-KO and DKO mice regained liver weight by 14 days after PHX. The deletion of cMyc did not affect liver regeneration, which was similar to the WT mice. WT and cMyc-KO mice started regaining liver weight as early as 24 hours after PHX, with a peak proliferation response at 48 hours after PHX. HNF4α- KO and DKO showed a delayed response with liver weight increase by day 7 after PHX. The overall hepatocyte proliferation response by DKO mice following PHX was lower than that of other genotypes. Interestingly, the surviving HNF4α-KO and DKO mice showed re-expression of HNF4α at mRNA and protein levels on day 14 after PHX. This was accompanied by a significant increase in the expression of Krt19 and Epcam, hepatic progenitor cell markers, in the DKO mice on day 14 after PHX. Conclusion: These data indicate that, in the absence of HNF4α, cMyc contributes to hepatocyte-driven proliferation to compensate for the lost tissue mass. Furthermore, in the absence of both HNF4α and cMyc, HPC-driven proliferation occurs to support liver regeneration.


Subject(s)
Hepatectomy , Hepatocyte Nuclear Factor 4 , Liver Regeneration , Mice, Knockout , Animals , Liver Regeneration/physiology , Hepatocyte Nuclear Factor 4/metabolism , Hepatocyte Nuclear Factor 4/genetics , Mice , Cell Proliferation , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Hepatocytes/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL
7.
J Nanobiotechnology ; 22(1): 521, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39210346

ABSTRACT

Tissue-derived extracellular vesicles (EVs) are emerging as pivotal players to maintain organ homeostasis, which show promise as a next-generation candidate for medical use with extensive source. However, the detailed function and therapeutic potential of tissue EVs remain insufficiently studied. Here, through bulk and single-cell RNA sequencing analyses combined with ultrastructural tissue examinations, we first reveal that in situ liver tissue EVs (LT-EVs) contribute to the intricate liver regenerative process after partial hepatectomy (PHx), and that hepatocytes are the primary source of tissue EVs in the regenerating liver. Nanoscale and proteomic profiling further identify that the hepatocyte-specific tissue EVs (Hep-EVs) are strengthened to release with carrying proliferative messages after PHx. Moreover, targeted inhibition of Hep-EV release via AAV-shRab27a in vivo confirms that Hep-EVs are required to orchestrate liver regeneration. Mechanistically, Hep-EVs from the regenerating liver reciprocally stimulate hepatocyte proliferation by promoting cell cycle progression through Cyclin-dependent kinase 1 (Cdk1) activity. Notably, supplementing with Hep-EVs from the regenerating liver demonstrates translational potential and ameliorates insufficient liver regeneration. This study provides a functional and mechanistic framework showing that the release of regenerative Hep-EVs governs rapid liver regeneration, thereby enriching our understanding of physiological and endogenous tissue EVs in organ regeneration and therapy.


Subject(s)
Cell Proliferation , Extracellular Vesicles , Hepatectomy , Hepatocytes , Liver Regeneration , Liver , Liver Regeneration/physiology , Extracellular Vesicles/metabolism , Hepatocytes/metabolism , Animals , Liver/metabolism , Mice , Humans , Male , Mice, Inbred C57BL , Regenerative Medicine/methods , CDC2 Protein Kinase/metabolism , Proteomics
8.
In Vivo ; 38(5): 2261-2270, 2024.
Article in English | MEDLINE | ID: mdl-39187322

ABSTRACT

BACKGROUND/AIM: The liver effectively restores both size and function following partial hepatectomy (PHx). Angiogenesis is crucial for the repair and regeneration of liver tissue post-PHx. Calcitonin gene-related peptide (CGRP) released from sensory nerves and its receptor-receptor activity-modifying protein 1 (RAMP1) are involved in angiogenesis. This study aimed to assess the role of RAMP1 signaling in angiogenesis during liver regeneration following PHx. MATERIALS AND METHODS: RAMP1 deficient (RAMP1-/-) and wild-type (WT) mice were subjected to PHx. RESULTS: RAMP1-/- mice demonstrated delayed liver regeneration, indicated by lower liver-to-body weight ratios compared to WT mice. This was associated with lower levels of Ki67+ hepatocytes and hepatic trophic growth factors. Additionally, RAMP1-/- mice exhibited lower levels of endothelial cell markers, including CD31, compared to WT mice. This reduction was associated with reduced levels of vascular endothelial growth factor (VEGF)-C, VEGF-D, and VEGF receptor 3 (VEGFR3). In WT mice with PHx, the administration of a VEGFR3 inhibitor reduced the liver-to-body weight ratio, Ki67+ hepatocytes, and VEGF-C/VEGFR3 expression levels in the liver compared to those in the vehicle-treated group. CONCLUSION: The deletion of RAMP1 signaling suppresses liver regeneration and angiogenesis through VEGFR3. Specific activation of RAMP1 signaling may represent a potential therapeutic strategy for liver regeneration following PHx.


Subject(s)
Hepatectomy , Liver Regeneration , Mice, Knockout , Neovascularization, Physiologic , Receptor Activity-Modifying Protein 1 , Signal Transduction , Animals , Liver Regeneration/physiology , Hepatectomy/methods , Mice , Receptor Activity-Modifying Protein 1/metabolism , Receptor Activity-Modifying Protein 1/genetics , Liver/metabolism , Liver/blood supply , Liver/surgery , Hepatocytes/metabolism , Male , Angiogenesis
9.
Endocrinology ; 165(8)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38963813

ABSTRACT

Vitamin D signals through the vitamin D receptor (VDR) to induce its end-organ effects. Hepatic stellate cells control development of liver fibrosis in response to stressors and vitamin D signaling decreases fibrogenesis. VDR expression in hepatocytes is low in healthy liver, and the role of VDR in hepatocyte proliferation is unclear. Hepatocyte-VDR null mice (hVDR) were used to assess the role of VDR and vitamin D signaling in hepatic regeneration. hVDR mice have impaired liver regeneration and impaired hepatocyte proliferation associated with significant differential changes in bile salts. Notably, mice lacking hepatocyte VDR had significant increases in expression of conjugated bile acids after partial hepatectomy, consistent with failure to normalize hepatic function by the 14-day time point tested. Real-time PCR of hVDR and control livers showed significant changes in expression of cell-cycle genes including cyclins D1 and E1 and cyclin-dependent kinase 2. Gene expression profiling of hepatocytes treated with vitamin D or control showed regulation of groups of genes involved in liver proliferation, hepatitis, liver hyperplasia/hyperproliferation, and liver necrosis/cell death. Together, these studies demonstrate an important functional role for VDR in hepatocytes during liver regeneration. Combined with the known profibrotic effects of impaired VDR signaling in stellate cells, the studies provide a mechanism whereby vitamin D deficiency would both reduce hepatocyte proliferation and permit fibrosis, leading to significant liver compromise.


Subject(s)
Bile Acids and Salts , Cell Proliferation , Hepatectomy , Hepatocytes , Liver Regeneration , Mice, Knockout , Receptors, Calcitriol , Animals , Liver Regeneration/drug effects , Liver Regeneration/physiology , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Male , Mice , Hepatocytes/metabolism , Hepatocytes/drug effects , Cell Proliferation/drug effects , Bile Acids and Salts/metabolism , Liver/metabolism , Cyclin D1/metabolism , Cyclin D1/genetics , Cyclin E/metabolism , Cyclin E/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 2/genetics , Mice, Inbred C57BL , Vitamin D/pharmacology , Signal Transduction/drug effects , Oncogene Proteins
10.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38975841

ABSTRACT

The liver is a remarkable organ that can regenerate in response to injury. Depending on the extent of injury, the liver can undergo compensatory hyperplasia or fibrosis. Despite decades of research, the molecular mechanisms underlying these processes are poorly understood. Here, we developed a new model to study liver regeneration based on cryoinjury. To visualise liver regeneration at cellular resolution, we adapted the CUBIC tissue-clearing approach. Hepatic cryoinjury induced a localised necrotic and apoptotic lesion characterised by inflammation and infiltration of innate immune cells. After this initial phase, we observed fibrosis, which resolved as regeneration re-established homeostasis in 30 days. Importantly, this approach enables the comparison of healthy and injured parenchyma within an individual animal, providing unique advantages to previous models. In summary, the hepatic cryoinjury model provides a fast and reproducible method for studying the cellular and molecular pathways underpinning fibrosis and liver regeneration.


Subject(s)
Liver Regeneration , Liver , Liver Regeneration/physiology , Animals , Liver/pathology , Liver/metabolism , Mice , Disease Models, Animal , Apoptosis , Mice, Inbred C57BL , Male , Liver Cirrhosis/pathology
11.
Ulus Travma Acil Cerrahi Derg ; 30(7): 472-479, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967532

ABSTRACT

BACKGROUND: Traumatic liver injury is an acute event that triggers liver repair. The augmenter of liver regeneration (ALR) has been identified as a growth factor involved in this process. This study evaluates the impact of ALR on isolated liver blunt trauma and examines its relationship with various time intervals. METHODS: Forty healthy female Wistar albino rats were divided into five groups (n=8 each). Isolated blunt liver trauma was induced using a custom-designed trauma platform in all groups except for Group 1. The groups were categorized by the timing of euthanasia post-trauma: 2nd (15 minutes), 3rd (30 minutes), 4th (45 minutes), and 5th (60 minutes). Assessments included plasma ALR levels, liver tissue ALR levels (both intact and lacerated), biochemical indices, and liver histological analysis. RESULTS: Plasma ALR levels in Group 4 were higher than in Groups 1 and 2 (p<0.01). Intact liver ALR levels in Groups 3 and 4 exceeded those in Group 1 (p<0.05, p<0.01, respectively). Intact liver tissue ALR levels in Group 5 were lower than in Groups 3 and 4 (p<0.05, p<0.01, respectively). Lacerated liver tissue ALR levels in Group 5 were higher than those in Groups 2 and 3. In Group 1, the plasma ALR level was higher than the intact liver tissue ALR level (p<0.05). In Group 2, plasma ALR levels exceeded those in intact liver tissue ALR levels (p<0.01). In Group 3, plasma ALR levels surpassed both lacerated and intact liver tissue ALR levels (p<0.05, p<0.001, respectively). In Group 4, the plasma ALR level was higher than the intact liver tissue ALR level (p<0.01), and the lacerated liver tissue level was higher than the intact liver ALR level (p<0.001). Additionally, inflammation scores were higher in Groups 3, 4, and 5 compared to Group 2 (p<0.05, p<0.01, p<0.01, respectively). CONCLUSION: This study is the first to explore the role of ALR in isolated blunt liver trauma. Following blunt liver trauma, both plasma and liver tissue ALR levels change within minutes.


Subject(s)
Disease Models, Animal , Liver Regeneration , Liver , Rats, Wistar , Wounds, Nonpenetrating , Animals , Female , Liver/injuries , Rats , Wounds, Nonpenetrating/pathology , Wounds, Nonpenetrating/complications , Liver Regeneration/physiology , Intercellular Signaling Peptides and Proteins/blood , Oxidoreductases Acting on Sulfur Group Donors
12.
Life Sci ; 353: 122924, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39038511

ABSTRACT

The liver is considered unique in its enormous capacity for regeneration and self-repair. In contrast to other regenerative organs (i.e., skin, skeletal muscle, and intestine), whether the adult liver contains a defined department of stem cells is still controversial. In order to compensate for the massive loss of hepatocytes following liver injury, the liver processes a precisely controlled transcriptional reprogram that can trigger cell proliferation and cell-fate switch. Epigenetic events are thought to regulate the organization of chromatin architecture and gene transcription during the liver regenerative process. In this review, we will summarize how changes to the chromatin by epigenetic modifiers are translated into cell fate transitions to restore liver homeostasis during liver regeneration.


Subject(s)
Epigenesis, Genetic , Liver Regeneration , Liver Regeneration/genetics , Liver Regeneration/physiology , Humans , Animals , Liver/metabolism , Liver/physiology , Hepatocytes/metabolism , Hepatocytes/physiology , Cell Proliferation/genetics
13.
Biochem Pharmacol ; 227: 116437, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025410

ABSTRACT

The normal liver has an extraordinary capacity of regeneration. However, this capacity is significantly impaired in steatotic livers. Emerging evidence indicates that metabolic dysfunction associated steatotic liver disease (MASLD) and liver regeneration share several key mechanisms. Some classical liver regeneration pathways, such as HGF/c-Met, EGFR, Wnt/ß-catenin and Hippo/YAP-TAZ are affected in MASLD. Some recently established therapeutic targets for MASH such as the Thyroid Hormone (TH) receptors, Glucagon-like protein 1 (GLP1), Farnesoid X receptor (FXR), Peroxisome Proliferator-Activated Receptors (PPARs) as well as Fibroblast Growth Factor 21 (FGF21) are also reported to affect hepatocyte proliferation. With this review we aim to provide insight into common molecular pathways, that may ultimately enable therapeutic strategies that synergistically ameliorate steatohepatitis and improve the regenerating capacity of steatotic livers. With the recent rise of prolonged ex-vivo normothermic liver perfusion prior to organ transplantation such treatment is no longer restricted to patients undergoing major liver resection or transplantation, but may eventually include perfused (steatotic) donor livers or even liver segments, opening hitherto unexplored therapeutic avenues.


Subject(s)
Fatty Liver , Liver Regeneration , Humans , Liver Regeneration/physiology , Animals , Fatty Liver/metabolism
14.
Nat Commun ; 15(1): 5827, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992008

ABSTRACT

The liver has the remarkable capacity to regenerate. In the clinic, regeneration is induced by portal vein embolization, which redirects portal blood flow, resulting in liver hypertrophy in locations with increased blood supply, and atrophy of embolized segments. Here, we apply single-cell and single-nucleus transcriptomics on healthy, hypertrophied, and atrophied patient-derived liver samples to explore cell states in the regenerating liver. Our data unveils pervasive upregulation of genes associated with developmental processes, cellular adhesion, and inflammation in post-portal vein embolization liver, disrupted portal-central hepatocyte zonation, and altered cell subtype composition of endothelial and immune cells. Interlineage crosstalk analysis reveals mesenchymal cells as an interaction hub between immune and endothelial cells, and highlights the importance of extracellular matrix proteins in liver regeneration. Moreover, we establish tissue-scale iterative indirect immunofluorescence imaging for high-dimensional spatial analysis of perivascular microenvironments, uncovering changes to tissue architecture in regenerating liver lobules. Altogether, our data is a rich resource revealing cellular and histological changes in human liver regeneration.


Subject(s)
Embolization, Therapeutic , Liver Regeneration , Liver , Portal Vein , Humans , Liver Regeneration/physiology , Embolization, Therapeutic/methods , Hepatocytes/metabolism , Single-Cell Analysis , Transcriptome , Male , Endothelial Cells/metabolism , Female , Hypertrophy , Middle Aged
15.
Pathol Res Pract ; 260: 155452, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972165

ABSTRACT

The development of amoebic liver abscess (ALA) leads to liver necrosis, accompanied by an exacerbated inflammatory response and the formation of multiple granulomas. Adequate management of the infection through the administration of treatment and the timely response of the organ to the damage allows the injury to heal with optimal regeneration without leaving scar tissue, which does not occur in other types of damage such as viral hepatitis that may conducts to fibrosis or cirrhosis. The Hedgehog signaling pathway (Hh) is crucial in the embryonic stage, while in adults it is usually reactivated in response to acute or chronic injuries, regeneration, and wound healing. In this work, we characterized Hh in experimental hepatic amoebiasis model, with the administration of treatment with metronidazole, as well as a pathway inhibitor (cyclopamine), through histological and immunohistochemical analyses including an ultrastructure analysis through transmission electron microscopy. The results showed an increase in the percentage of lesions obtained, a decrease in the presence of newly formed hepatocytes, a generalized inflammatory response, irregular distribution of type I collagen accompanied by the presence of fibroblast-type cells and a decrease in effector cells of this pathway. These results constitute the first evidence of the association of the activation of Hh with the liver regeneration process in experimental amebiasis.


Subject(s)
Disease Models, Animal , Hedgehog Proteins , Liver Regeneration , Signal Transduction , Liver Regeneration/physiology , Hedgehog Proteins/metabolism , Animals , Liver Abscess, Amebic/pathology , Male , Liver/pathology , Liver/metabolism , Metronidazole/pharmacology , Metronidazole/therapeutic use , Veratrum Alkaloids/pharmacology
16.
J Pharm Biomed Anal ; 249: 116369, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39047463

ABSTRACT

Accurate assessment of future liver remnant growth after partial hepatectomy (PH) in patients with different liver backgrounds is a pressing clinical issue. Amino acid (AA) metabolism plays a crucial role in liver regeneration. In this study, we combined metabolomics and machine learning (ML) to develop a generalized future liver remnant assessment model for multiple liver backgrounds. The liver index was calculated at 0, 6, 24, 48, 72 and 168 h after 70 % PH in healthy mice and mice with nonalcoholic steatohepatitis or liver fibrosis. The serum levels of 39 amino acids (AAs) were measured using UPLC-MS/MS. The dataset was randomly divided into training and testing sets at a 2:1 ratio, and orthogonal partial least squares regression (OPLS) and minimally biased variable selection in R (MUVR) were used to select a metabolite signature of AAs. To assess liver remnant growth, nine ML models were built, and evaluated using the coefficient of determination (R2), mean absolute error (MAE), and root mean square error (RMSE). The post-Pareto technique for order preference by similarity to the ideal solution (TOPSIS) was employed for ranking the ML algorithms, and a stacking technique was utilized to establish consensus among the superior algorithms. Compared with those of OPLS, the signature AAs set identified by MUVR (Thr, Arg, EtN, Phe, Asa, 3MHis, Abu, Asp, Tyr, Leu, Ser, and bAib) are more concise. Post-Pareto TOPSIS ranking demonstrated that the majority of ML algorithm in combinations with MUVR outperformed those with OPLS. The established SVM-KNN consensus model performed best, with an R2 of 0.79, an MAE of 0.0029, and an RMSE of 0.0035 for the testing set. This study identified a metabolite signature of 12 AAs and constructed an SVM-KNN consensus model to assess future liver remnant growth after PH in mice with different liver backgrounds. Our preclinical study is anticipated to establish an alternative and generalized assessment method for liver regeneration.


Subject(s)
Amino Acids , Hepatectomy , Liver Regeneration , Liver , Machine Learning , Metabolomics , Tandem Mass Spectrometry , Animals , Hepatectomy/methods , Metabolomics/methods , Mice , Liver/metabolism , Liver/surgery , Amino Acids/metabolism , Amino Acids/blood , Liver Regeneration/physiology , Male , Tandem Mass Spectrometry/methods , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/surgery , Liver Cirrhosis/surgery , Liver Cirrhosis/metabolism , Disease Models, Animal , Chromatography, High Pressure Liquid/methods
17.
Adv Sci (Weinh) ; 11(32): e2402856, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38923873

ABSTRACT

Lack of liver regenerative capacity is the primary cause of hepatic failure and even mortality in patients undergoing hepatectomy, with no effective intervention strategies currently available. Therefore, identifying efficacious interventions to enhance liver regeneration is pivotal for optimizing clinical outcomes. Recent studies have demonstrated that vagotomy exerts an inhibitory effect on liver regeneration following partial hepatectomy, thereby substantiating the pivotal role played by the vagus nerve in the process of liver regeneration. In recent years, electroacupuncture (EA) has emerged as a non-invasive technique for stimulating the vagus nerve. However, EA on hepatic regeneration remains uncertain. In this study, a 70% partial hepatectomy (PH) mouse model is utilized to investigate the effects of EA on acute liver regeneration and elucidate its underlying molecular mechanisms. It is observed that EA at ST36 acutely activated cholinergic neurons in the dorsal motor nucleus of the vagus nerve (DMV), resulting in increased release of acetylcholine from hepatic vagal nerve endings and subsequent activation of IL-6 signaling in liver macrophages. Ultimately, these events promoted hepatocyte proliferation and facilitated liver regeneration. These findings provide insights into the fundamental brain-liver axis mechanism through which EA promotes liver regeneration, offering a novel therapeutic approach for post-hepatectomy liver regeneration disorders.


Subject(s)
Disease Models, Animal , Electroacupuncture , Hepatectomy , Liver Regeneration , Macrophages , Vagus Nerve , Animals , Electroacupuncture/methods , Hepatectomy/methods , Liver Regeneration/physiology , Mice , Vagus Nerve/surgery , Vagus Nerve/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Liver/surgery , Liver/metabolism
18.
J Vis Exp ; (207)2024 May 24.
Article in English | MEDLINE | ID: mdl-38856217

ABSTRACT

Partial 2/3 hepatectomy in mice is used in research to study the liver's regenerative capacity and explore outcomes of liver resection in a number of disease models. In the classical partial 2/3 hepatectomy in mice, two of the five liver lobes, namely the left and median lobes representing approximately 66% of the liver mass, are resected en bloc with an expected postoperative survival of 100%. More aggressive partial hepatectomies are technically more challenging and hence, have seldom been used in mice. Our group has developed a mouse model of an extended hepatectomy technique in which three of the five liver lobes, including the left, median, and right upper lobes, are resected separately to remove approximately 78% of the total liver mass. This extended resection, in otherwise healthy mice, leaves a remnant liver that cannot always sustain adequate and timely regeneration. Failure to regenerate ultimately results in 50% postoperative lethality within 1 week due to fulminant hepatic failure. This procedure of extended 78% hepatectomy in mice represents a unique surgical model for the study of small-for-size syndrome and the evaluation of therapeutic strategies to improve liver regeneration and outcomes in the setting of liver transplantation or extended liver resection for cancer.


Subject(s)
Hepatectomy , Liver Regeneration , Models, Animal , Animals , Hepatectomy/methods , Mice , Liver Regeneration/physiology , Liver/surgery , Disease Models, Animal
19.
Hepatol Commun ; 8(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38836805

ABSTRACT

BACKGROUND: Extended liver resection is the only treatment option for perihilar cholangiocarcinoma (pCCA). Bile salts and the gut hormone FGF19, both promoters of liver regeneration (LR), have not been investigated in patients undergoing resection for pCCA. We aimed to evaluate the bile salt-FGF19 axis perioperatively in pCCA and study its effects on LR. METHODS: Plasma bile salts, FGF19, and C4 (bile salt synthesis marker) were assessed in patients with pCCA and controls (colorectal liver metastases), before and after resection on postoperative days (PODs) 1, 3, and 7. Hepatic bile salts were determined in intraoperative liver biopsies. RESULTS: Partial liver resection in pCCA elicited a sharp decline in bile salt and FGF19 plasma levels on POD 1 and remained low thereafter, unlike in controls, where bile salts rose gradually. Preoperatively, suppressed C4 in pCCA normalized postoperatively to levels similar to those in the controls. The remnant liver volume and postoperative bilirubin levels were negatively associated with postoperative C4 levels. Furthermore, patients who developed postoperative liver failure had nearly undetectable C4 levels on POD 7. Hepatic bile salts strongly predicted hyperbilirubinemia on POD 7 in both groups. Finally, postoperative bile salt levels on day 7 were an independent predictor of LR. CONCLUSIONS: Partial liver resection alters the bile salt-FGF19 axis, but its derailment is unrelated to LR in pCCA. Postoperative monitoring of circulating bile salts and their production may be useful for monitoring LR.


Subject(s)
Bile Acids and Salts , Bile Duct Neoplasms , Fibroblast Growth Factors , Hepatectomy , Klatskin Tumor , Liver Regeneration , Humans , Male , Bile Acids and Salts/blood , Bile Acids and Salts/metabolism , Fibroblast Growth Factors/blood , Bile Duct Neoplasms/surgery , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/blood , Female , Klatskin Tumor/surgery , Klatskin Tumor/pathology , Klatskin Tumor/blood , Middle Aged , Liver Regeneration/physiology , Aged , Case-Control Studies , Liver/metabolism , Liver/surgery
20.
Biochem Pharmacol ; 226: 116377, 2024 08.
Article in English | MEDLINE | ID: mdl-38906228

ABSTRACT

BTB and CNC homology 1 (BACH1) regulates biological processes, including energy metabolism and oxidative stress. Insufficient liver regeneration after hepatectomy remains an issue for surgeons. The Pringle maneuver is widely used during hepatectomy and induces ischemia/reperfusion (I/R) injury in hepatocytes. A rat model of two-thirds partial hepatectomy with repeated I/R treatment was used to simulate clinical hepatectomy with Pringle maneuver. Delayed recovery of liver function after hepatectomy with the repeated Pringle maneuver in clinic and impaired liver regeneration in rat model were observed. Highly elevated lactate levels, along with reduced mitochondrial complex III and IV activities in liver tissues, indicated that the glycolytic phenotype was promoted after hepatectomy with repeated I/R. mRNA expression profile analysis of glycolysis-related genes in clinical samples and further verification experiments in rat models showed that high BACH1 expression levels correlated with the glycolytic phenotype after hepatectomy with repeated I/R. BACH1 overexpression restricted the proliferative potential of hepatocytes stimulated with HGF. High PDK1 expression and high lactate levels, together with low mitochondrial complex III and IV activities and reduced ATP concentrations, were detected in BACH1-overexpressing hepatocytes with HGF stimulation. Moreover, HO-1 expression was downregulated, and oxidative stress was exacerbated in the BACH1-overexpressing hepatocytes with HGF stimulation. Cell experiments involving repeated hypoxia/reoxygenation revealed that reactive oxygen species accumulation triggered the TGF-ß1/BACH1 axis in hepatocytes. Finally, inhibiting BACH1 with the inhibitor hemin effectively restored the liver regenerative ability after hepatectomy with repeated I/R. These results provide a potential therapeutic strategy for impaired liver regeneration after repeated I/R injury.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Energy Metabolism , Hepatectomy , Hepatocytes , Liver Regeneration , Oxidative Stress , Rats, Sprague-Dawley , Reperfusion Injury , Animals , Humans , Male , Rats , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Energy Metabolism/physiology , Hepatocytes/metabolism , Liver Regeneration/physiology , Oxidative Stress/physiology , Reperfusion Injury/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL