Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.053
Filter
1.
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
3.
Basic Clin Pharmacol Toxicol ; 135(2): 148-163, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38887973

ABSTRACT

Human pregnane X receptor (PXR) is critical for regulating the expression of key drug-metabolizing enzymes such as CYP3A and CYP2C. Our recent study revealed that treatment with rodent-specific PXR agonist pregnenolone-16α-carbonitrile (PCN) significantly induced hepatomegaly and promoted liver regeneration after two-thirds partial hepatectomy (PHx) in mice. However, it remains unclear whether PXR activation induces hepatomegaly and liver regeneration and simultaneously promotes metabolic function of the liver. Here, we investigated the metabolism activity of CYP1A2, CYP3A1/2 and CYP2C6/11 during PXR activation-induced liver enlargement and regeneration in rats after cocktail dosing of CYP probe drugs. For PCN-induced hepatomegaly, a notable increase in the metabolic activity of CYP3A1/2 and CYP2C6/11, as evidenced by the plasma exposure of probe substrates and the AUC ratios of the characteristic metabolites to its corresponding probe substrates. The metabolic activity of CYP1A2, CYP3A1/2 and CYP2C6/11 decreased significantly after PHx. However, PCN treatment obviously enhanced the metabolic activity of CYP2C6/11 and CYP3A1/2 in PHx rats. Furthermore, the protein expression levels of CYP3A1/2 and CYP2C6/11 in liver were up-regulated. Taken together, this study demonstrates that PXR activation not only induces hepatomegaly and liver regeneration in rats, but also promotes the protein expression and metabolic activity of the PXR downstream metabolizing enzymes such as CYP3A1/2 and CYP2C6/11 in the body.


Subject(s)
Cytochrome P-450 CYP3A , Hepatomegaly , Liver Regeneration , Liver , Pregnane X Receptor , Pregnenolone Carbonitrile , Animals , Pregnane X Receptor/metabolism , Pregnane X Receptor/genetics , Liver Regeneration/drug effects , Male , Cytochrome P-450 CYP3A/metabolism , Pregnenolone Carbonitrile/pharmacology , Liver/metabolism , Liver/enzymology , Liver/drug effects , Rats , Hepatomegaly/metabolism , Hepatomegaly/pathology , Aryl Hydrocarbon Hydroxylases/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P450 Family 2/metabolism , Cytochrome P450 Family 2/genetics , Rats, Sprague-Dawley , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2/genetics , Steroid 16-alpha-Hydroxylase/metabolism , Steroid 16-alpha-Hydroxylase/genetics , Steroid 12-alpha-Hydroxylase/metabolism , Steroid 12-alpha-Hydroxylase/genetics , Hepatectomy
4.
Nature ; 630(8015): 158-165, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38693268

ABSTRACT

The liver has a unique ability to regenerate1,2; however, in the setting of acute liver failure (ALF), this regenerative capacity is often overwhelmed, leaving emergency liver transplantation as the only curative option3-5. Here, to advance understanding of human liver regeneration, we use paired single-nucleus RNA sequencing combined with spatial profiling of healthy and ALF explant human livers to generate a single-cell, pan-lineage atlas of human liver regeneration. We uncover a novel ANXA2+ migratory hepatocyte subpopulation, which emerges during human liver regeneration, and a corollary subpopulation in a mouse model of acetaminophen (APAP)-induced liver regeneration. Interrogation of necrotic wound closure and hepatocyte proliferation across multiple timepoints following APAP-induced liver injury in mice demonstrates that wound closure precedes hepatocyte proliferation. Four-dimensional intravital imaging of APAP-induced mouse liver injury identifies motile hepatocytes at the edge of the necrotic area, enabling collective migration of the hepatocyte sheet to effect wound closure. Depletion of hepatocyte ANXA2 reduces hepatocyte growth factor-induced human and mouse hepatocyte migration in vitro, and abrogates necrotic wound closure following APAP-induced mouse liver injury. Together, our work dissects unanticipated aspects of liver regeneration, demonstrating an uncoupling of wound closure and hepatocyte proliferation and uncovering a novel migratory hepatocyte subpopulation that mediates wound closure following liver injury. Therapies designed to promote rapid reconstitution of normal hepatic microarchitecture and reparation of the gut-liver barrier may advance new areas of therapeutic discovery in regenerative medicine.


Subject(s)
Liver Failure, Acute , Liver Regeneration , Animals , Female , Humans , Male , Mice , Acetaminophen/pharmacology , Cell Lineage , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/cytology , Liver/drug effects , Liver/pathology , Liver Failure, Acute/pathology , Liver Failure, Acute/chemically induced , Liver Regeneration/drug effects , Mice, Inbred C57BL , Necrosis/chemically induced , Regenerative Medicine , Single-Cell Gene Expression Analysis , Wound Healing
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167166, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642480

ABSTRACT

BACKGROUND AND AIMS: Liver regeneration is essential for the preservation of homeostasis and survival. Bile acids (BAs)-mediated signaling is necessary for liver regeneration, but BAs levels need to be carefully controlled to avoid hepatotoxicity. We studied the early response of the BAs-fibroblast growth factor 19 (FGF19) axis in healthy individuals undergoing hepatectomy for living donor liver transplant. We also evaluated BAs synthesis in mice upon partial hepatectomy (PH) and acute inflammation, focusing on the regulation of cytochrome-7A1 (CYP7A1), a key enzyme in BAs synthesis from cholesterol. METHODS: Serum was obtained from twelve human liver donors. Mice underwent 2/3-PH or sham-operation. Acute inflammation was induced with bacterial lipopolysaccharide (LPS) in mice fed control or antoxidant-supplemented diets. BAs and 7α-hydroxy-4-cholesten-3-one (C4) levels were measured by HPLC-MS/MS; serum FGF19 by ELISA. Gene expression and protein levels were analyzed by RT-qPCR and western-blot. RESULTS: Serum BAs levels increased after PH. In patients with more pronounced hypercholanemia, FGF19 concentrations transiently rose, while C4 levels (a readout of CYP7A1 activity) dropped 2 h post-resection in all cases. Serum BAs and C4 followed the same pattern in mice 1 h after PH, but C4 levels also dropped in sham-operated and LPS-treated animals, without marked changes in CYP7A1 protein levels. LPS-induced serum C4 decline was attenuated in mice fed an antioxidant-supplemented diet. CONCLUSIONS: In human liver regeneration FGF19 upregulation may constitute a protective response from BAs excess during liver regeneration. Our findings suggest the existence of post-translational mechanisms regulating CYP7A1 activity, and therefore BAs synthesis, independent from CYP7A1/Cyp7a1 gene transcription.


Subject(s)
Bile Acids and Salts , Cholesterol 7-alpha-Hydroxylase , Fibroblast Growth Factors , Hepatectomy , Liver Regeneration , Humans , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/biosynthesis , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Liver Regeneration/drug effects , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Mice , Male , Female , Adult , Middle Aged , Liver/metabolism , Mice, Inbred C57BL , Liver Transplantation , Lipopolysaccharides/pharmacology
8.
Toxicology ; 504: 153804, 2024 May.
Article in English | MEDLINE | ID: mdl-38614205

ABSTRACT

Fifty percent of all acute liver failure (ALF) cases in the United States are due to acetaminophen (APAP) overdose. Assessment of canonical features of liver injury, such as plasma alanine aminotransferase activities are poor predictors of acute liver failure (ALF), suggesting the involvement of additional mechanisms independent of hepatocyte death. Previous work demonstrated a severe overdose of APAP results in impaired regeneration, the induction of senescence by p21, and increased mortality. We hypothesized that a discrete population of p21+ hepatocytes acquired a secretory phenotype that directly impedes liver recovery after a severe APAP overdose. Leveraging in-house human APAP explant liver and publicly available single-nuclei RNAseq data, we identified a subpopulation of p21+ hepatocytes enriched in a unique secretome of factors, such as CXCL14. Spatial transcriptomics in the mouse model of APAP overdose confirmed the presence of a p21+ hepatocyte population that directly surrounded the necrotic areas. In both male and female mice, we found a dose-dependent induction of p21 and persistent circulating levels of the p21-specific constituent, CXCL14, in the plasma after a severe APAP overdose. In parallel experiments, we targeted either the putative senescent hepatocytes with the senolytic drugs, dasatinib and quercetin, or CXCL14 with a neutralizing antibody. We found that targeting CXCL14 greatly enhanced liver recovery after APAP-induced liver injury, while targeting senescent hepatocytes had no effect. These data support the conclusion that the sustained induction of p21 in hepatocytes with persistent CXCL14 secretion are critical mechanistic events leading to ALF in mice and human patients.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Chemokines, CXC , Cyclin-Dependent Kinase Inhibitor p21 , Hepatocytes , Mice, Inbred C57BL , Acetaminophen/toxicity , Animals , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Male , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Mice , Chemokines, CXC/metabolism , Chemokines, CXC/genetics , Liver Regeneration/drug effects , Drug Overdose , Analgesics, Non-Narcotic/toxicity
9.
Adv Healthc Mater ; 13(17): e2303828, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608209

ABSTRACT

Partial hepatectomy is an essential surgical technique used to treat advanced liver diseases such as liver tumors, as well as for performing liver transplants from living donors. However, postoperative complications such as bleeding, abdominal adhesions, wound infections, and inadequate liver regeneration pose significant challenges and increase morbidity and mortality rates. A self-repairing mixed hydrogel (O5H2/Cu2+/SCCK), containing stem cell derived cytokine (SCCK) derived from human umbilical cord mesenchymal stem cells (HUMSCs) treated with the traditional Chinese remedy Tanshinone IIA (TSA), is developed. This SCCK, in conjunction with O5H2, demonstrates remarkable effects on Kupffer cell activation and extracellular matrix (ECM) remodeling. This leads to the secretion of critical growth factors promoting enhanced proliferation of hepatocytes and endothelial cells, thereby facilitating liver regeneration and repair after partial hepatectomy. Furthermore, the hydrogel, featuring macrophage-regulating properties, effectively mitigates inflammation and oxidative stress damage in the incision area, creating an optimal environment for postoperative liver regeneration. The injectability and strong adhesion of the hydrogel enables rapid hemostasis at the incision site, while its physical barrier function prevents postoperative abdominal adhesions. Furthermore, the hydrogel's incorporation of Cu2+ provides comprehensive antibacterial effects, protecting against a wide range of bacteria types and reducing the chances of infections after surgery.


Subject(s)
Extracellular Matrix , Hepatectomy , Hydrogels , Kupffer Cells , Liver Regeneration , Liver Regeneration/drug effects , Liver Regeneration/physiology , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Humans , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Mice , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Male , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Cytokines/metabolism , Mice, Inbred C57BL
10.
Acta Pharmacol Sin ; 45(6): 1201-1213, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491160

ABSTRACT

The angiotensin II type 2 receptor (AT2R) is a well-established component of the renin-angiotensin system and is known to counteract classical activation of this system and protect against organ damage. Pharmacological activation of the AT2R has significant therapeutic benefits, including vasodilation, natriuresis, anti-inflammatory activity, and improved insulin sensitivity. However, the precise biological functions of the AT2R in maintaining homeostasis in liver tissue remain largely unexplored. In this study, we found that the AT2R facilitates liver repair and regeneration following acute injury by deactivating Hippo signaling and that interleukin-6 transcriptionally upregulates expression of the AT2R in hepatocytes through STAT3 acting as a transcription activator binding to promoter regions of the AT2R. Subsequently, elevated AT2R levels activate downstream signaling via heterotrimeric G protein Gα12/13-coupled signals to induce Yap activity, thereby contributing to repair and regeneration processes in the liver. Conversely, a deficiency in the AT2R attenuates regeneration of the liver while increasing susceptibility to acetaminophen-induced liver injury. Administration of an AT2R agonist significantly enhances the repair and regeneration capacity of injured liver tissue. Our findings suggest that the AT2R acts as an upstream regulator in the Hippo pathway and is a potential target in the treatment of liver damage.


Subject(s)
Hippo Signaling Pathway , Interleukin-6 , Liver Regeneration , Mice, Inbred C57BL , Protein Serine-Threonine Kinases , Receptor, Angiotensin, Type 2 , Signal Transduction , Animals , Male , Mice , Acetaminophen , Adaptor Proteins, Signal Transducing/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Interleukin-6/metabolism , Liver/metabolism , Liver/drug effects , Liver Regeneration/drug effects , Liver Regeneration/physiology , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , YAP-Signaling Proteins/metabolism
11.
Arch Toxicol ; 98(6): 1843-1858, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38551724

ABSTRACT

Acetaminophen (APAP)-induced hepatotoxicity is comprised of an injury and recovery phase. While pharmacological interventions, such as N-acetylcysteine (NAC) and 4-methylpyrazole (4-MP), prevent injury there are no therapeutics that promote recovery. JNJ-26366821 (TPOm) is a novel thrombopoietin mimetic peptide with no sequence homology to endogenous thrombopoietin (TPO). Endogenous thrombopoietin is produced by hepatocytes and the TPO receptor is present on liver sinusoidal endothelial cells in addition to megakaryocytes and platelets, and we hypothesize that TPOm activity at the TPO receptor in the liver provides a beneficial effect following liver injury. Therefore, we evaluated the extent to which TPOm, NAC or 4-MP can provide a protective and regenerative effect in the liver when administered 2 h after an APAP overdose of 300 mg/kg in fasted male C57BL/6J mice. TPOm did not affect protein adducts, oxidant stress, DNA fragmentation and hepatic necrosis up to 12 h after APAP. In contrast, TPOm treatment was beneficial at 24 h, i.e., all injury parameters were reduced by 42-48%. Importantly, TPOm enhanced proliferation by 100% as indicated by PCNA-positive hepatocytes around the area of necrosis. When TPOm treatment was delayed by 6 h, there was no effect on the injury, but a proliferative effect was still evident. In contrast, 4MP and NAC treated at 2 h after APAP significantly attenuated all injury parameters at 24 h but failed to enhance hepatocyte proliferation. Thus, TPOm arrests the progression of liver injury by 24 h after APAP and accelerates the onset of the proliferative response which is essential for liver recovery.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Liver Regeneration , Liver , Mice, Inbred C57BL , Thrombopoietin , Animals , Acetaminophen/toxicity , Male , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/drug therapy , Thrombopoietin/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Regeneration/drug effects , Mice , Acetylcysteine/pharmacology , Pyrazoles/pharmacology , Hepatocytes/drug effects , Oxidative Stress/drug effects , Receptors, Thrombopoietin/metabolism , Cell Proliferation/drug effects
12.
Proc Natl Acad Sci U S A ; 119(28): e2206113119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867764

ABSTRACT

The Hippo signaling pathway acts as a brake on regeneration in many tissues. This cascade of kinases culminates in the phosphorylation of the transcriptional cofactors Yap and Taz, whose concentration in the nucleus consequently remains low. Various types of cellular signals can reduce phosphorylation, however, resulting in the accumulation of Yap and Taz in the nucleus and subsequently in mitosis. We earlier identified a small molecule, TRULI, that blocks the final kinases in the pathway, Lats1 and Lats2, and thus elicits proliferation of several cell types that are ordinarily postmitotic and aids regeneration in mammals. In the present study, we present the results of chemical modification of the original compound and demonstrate that a derivative, TDI-011536, is an effective blocker of Lats kinases in vitro at nanomolar concentrations. The compound fosters extensive proliferation in retinal organoids derived from human induced pluripotent stem cells. Intraperitoneal administration of the substance to mice suppresses Yap phosphorylation for several hours and induces transcriptional activation of Yap target genes in the heart, liver, and skin. Moreover, the compound initiates the proliferation of cardiomyocytes in adult mice following cardiac cryolesions. After further chemical refinement, related compounds might prove useful in protective and regenerative therapies.


Subject(s)
Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Regeneration , Animals , Cell Proliferation/drug effects , Heart/physiology , Humans , Induced Pluripotent Stem Cells , Liver Regeneration/drug effects , Liver Regeneration/genetics , Liver Regeneration/physiology , Mice , Organoids/physiology , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Regeneration/drug effects , Regeneration/genetics , Retina/physiology , Skin Physiological Phenomena/drug effects , Skin Physiological Phenomena/genetics , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , YAP-Signaling Proteins/metabolism
13.
Naunyn Schmiedebergs Arch Pharmacol ; 395(3): 377-380, 2022 03.
Article in English | MEDLINE | ID: mdl-35076714

ABSTRACT

Almost every human organ has a poor ability to regenerate, notable exceptions are liver, skin, gut, etc. Molecular and cellular underpinnings of liver regeneration might pave the way for novel treatments concerned with chronic liver disorder. Such treatments would eliminate the disadvantages of liver transplantation, such as a scarcity of donor organs, a lengthy waitlist, significant medical expenses, surgical complications, and the necessity for lifelong immunosuppressive medications. Advancement in the development of regenerative therapy is giving hope to those suffering from end-stage liver disorder. The regeneration process is unique, intricate, and well coordinated, which involve the interaction of numerous signaling pathways, cytokines, and growth factor. Various signaling pathways for liver regeneration are HO-1/BER pathway, Tweak/Fn14 signaling pathway, Hippo pathway, Wnt/beta-catenin pathway, Hedgehog signaling pathway, bile acids repairing pathway, serotonin (5HT) pathway, estrogen pathway, thyrotropin-releasing hormone (TRH) pathway, insulin repairing pathway, etc. The in vitro scientific literature revealed that numerous GSK-3 ß inhibitors (LY 2090314, AR-A014418, Tideglusib, Solasodine, CHIR99021, 9-ING-41, SB-216763) play an important role in stimulating the liver regeneration process. Similarly, from the above discussion, the direction is highlighted to emphasize the proposed molecular Wnt/ß-catenin signaling pathway which is associated with GSK-3 ß inhibition for the induction of the repairing and regeneration process.


Subject(s)
End Stage Liver Disease/therapy , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Liver Regeneration/drug effects , Wnt Signaling Pathway/drug effects , Animals , Enzyme Inhibitors/pharmacology , Humans , Liver Regeneration/physiology , Signal Transduction/drug effects
14.
Hepatology ; 75(3): 584-599, 2022 03.
Article in English | MEDLINE | ID: mdl-34687050

ABSTRACT

BACKGROUND AND AIMS: The mechanisms involved in liver regeneration after partial hepatectomy (pHx) are complicated. Cellular senescence, once linked to aging, plays a pivotal role in wound repair. However, the regulatory effects of cellular senescence on liver regeneration have not been fully elucidated. APPROACH AND RESULTS: Mice subjected to pHx were analyzed 14 days after surgery. The incomplete remodeling of liver sinusoids affected shear stress-induced endothelial nitric oxide synthase (eNOS) signaling on day 14, resulting in the accumulation of senescent LSECs. Removing macrophages to augment LSEC senescence led to a malfunction of the regenerating liver. A dynamic fluctuation in Notch activity accompanied senescent LSEC accumulation during liver regeneration. Endothelial Notch activation by using Cdh5-CreERT NICeCA mice triggered LSEC senescence and senescence-associated secretory phenotype, which disrupted liver regeneration. Blocking the Notch by γ-secretase inhibitor (GSI) diminished senescence and promoted LSEC expansion. Mechanically, Notch-hairy and enhancer of split 1 signaling inhibited sirtuin 1 (Sirt1) transcription by binding to its promoter region. Activation of Sirt1 by SRT1720 neutralized the up-regulation of P53, P21, and P16 caused by Notch activation and eliminated Notch-driven LSEC senescence. Finally, Sirt1 activator promoted liver regeneration by abrogating LSEC senescence and improving sinusoid remodeling. CONCLUSIONS: Shear stress-induced LSEC senescence driven by Notch interferes with liver regeneration after pHx. Sirt1 inhibition accelerates liver regeneration by abrogating Notch-driven senescence, providing a potential opportunity to target senescent cells and facilitate liver repair after injury.


Subject(s)
Cellular Senescence , Liver Regeneration , Receptors, Notch , Signal Transduction/drug effects , Sirtuin 1/metabolism , Animals , Cellular Senescence/drug effects , Cellular Senescence/physiology , Gamma Secretase Inhibitors and Modulators/pharmacology , Hepatectomy/methods , Heterocyclic Compounds, 4 or More Rings/pharmacology , Liver Regeneration/drug effects , Liver Regeneration/physiology , Mice , Nitric Oxide Synthase Type III/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Senescence-Associated Secretory Phenotype/genetics
15.
Hepatology ; 75(1): 74-88, 2022 01.
Article in English | MEDLINE | ID: mdl-34387904

ABSTRACT

BACKGROUND AND AIMS: Peroxisome proliferator-activated receptor α (PPARα, NR1C1) is a ligand-activated nuclear receptor involved in the regulation of lipid catabolism and energy homeostasis. PPARα activation induces hepatomegaly and plays an important role in liver regeneration, but the underlying mechanisms remain unclear. APPROACH AND RESULTS: In this study, the effect of PPARα activation on liver enlargement and regeneration was investigated in several strains of genetically modified mice. PPARα activation by the specific agonist WY-14643 significantly induced hepatomegaly and accelerated liver regeneration after 70% partial hepatectomy (PHx) in wild-type mice and Pparafl/fl mice, while these effects were abolished in hepatocyte-specific Ppara-deficient (PparaΔHep ) mice. Moreover, PPARα activation promoted hepatocyte hypertrophy around the central vein area and hepatocyte proliferation around the portal vein area. Mechanistically, PPARα activation regulated expression of yes-associated protein (YAP) and its downstream targets (connective tissue growth factor, cysteine-rich angiogenic inducer 61, and ankyrin repeat domain 1) as well as proliferation-related proteins (cyclins A1, D1, and E1). Binding of YAP with the PPARα E domain was critical for the interaction between YAP and PPARα. PPARα activation further induced nuclear translocation of YAP. Disruption of the YAP-transcriptional enhancer factor domain family member (TEAD) association significantly suppressed PPARα-induced hepatomegaly and hepatocyte enlargement and proliferation. In addition, PPARα failed to induce hepatomegaly in adeno-associated virus-Yap short hairpin RNA-treated mice and liver-specific Yap-deficient mice. Blockade of YAP signaling abolished PPARα-induced hepatocyte hypertrophy around the central vein area and hepatocyte proliferation around the portal vein area. CONCLUSIONS: This study revealed a function of PPARα in regulating liver size and liver regeneration through activation of the YAP-TEAD signaling pathway. These findings have implications for understanding the physiological functions of PPARα and suggest its potential for manipulation of liver size and liver regeneration.


Subject(s)
Hepatomegaly/genetics , Liver Regeneration/genetics , PPAR alpha/metabolism , TEA Domain Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Hepatectomy/adverse effects , Hepatocytes/pathology , Hepatomegaly/pathology , Humans , Liver/pathology , Liver/surgery , Liver Regeneration/drug effects , Male , Mice , Mice, Transgenic , PPAR alpha/agonists , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Signal Transduction/genetics , YAP-Signaling Proteins/genetics
16.
J Pharmacol Sci ; 148(1): 19-30, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34924126

ABSTRACT

Hepatic ischemia/reperfusion (I/R) injury contributes to morbidity and mortality during liver resection or transplantation, with limited effective treatments available. Here, we investigated the potential benefits and underlying mechanisms of pterostilbene (Pt), a natural component of blueberries and grapes, in preventing hepatic I/R injury. Male C57BL/6 mice subjected to partial warm hepatic I/R and human hepatocyte cell line L02 cells exposed to anoxia/reoxygenation (A/R) were used as in vivo and in vitro models, respectively. Our findings showed that pretreatment with Pt ameliorated hepatic I/R injury by improving liver histology, decreasing hepatocyte apoptosis, and reducing plasma ALT and AST levels. Likewise, cell apoptosis, mitochondrial membrane dysfunction, and mitochondrial ROS overproduction in L02 cells triggered by the A/R challenge in vitro were reduced due to Pt administration. Mechanistically, Pt treatment efficiently enhanced mitophagy and upregulated PINK1, Parkin, and LC3B expression. Notably, the protective effect of Pt was largely abrogated after cells were transfected with PINK1 siRNA. Moreover, Pt pretreatment promoted hepatocyte proliferation and liver regeneration in the late phase of hepatic I/R. In conclusion, our findings provide evidence that Pt exerts hepatoprotective effects in hepatic I/R injury by upregulating PINK1-mediated mitophagy.


Subject(s)
Gene Expression Regulation/drug effects , Hepatic Infarction/genetics , Hepatic Infarction/prevention & control , Mitophagy/drug effects , Mitophagy/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/prevention & control , Stilbenes/pharmacology , Stilbenes/therapeutic use , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Hepatocytes/physiology , Humans , Liver Regeneration/drug effects , Male , Mice, Inbred C57BL , Up-Regulation/drug effects , Up-Regulation/genetics
17.
Acta cir. bras ; 37(9): e370901, 2022. tab, graf, ilus
Article in English | LILACS, VETINDEX | ID: biblio-1402981

ABSTRACT

Purpose: To evaluate the effect of preoperative intravenous chemotherapy with 5-fluorouracil on liver regeneration in an experimental model of major hepatectomy in rats. Methods: Wistar rats were divided into two groups of 20 animals each and submitted to 70% hepatectomy 24 h after intravenous injection of 5-fluorouracil 20 mg/kg (fluorouracil group, FG) or 0.9% saline (control group, CG). After hepatectomy, each group was subdivided into two subgroups of 10 animals each according to the day of sacrifice (24 h or 7 days). Liver weight during regeneration, liver regeneration rate using Kwon formula, and the immunohistochemical markers proliferating cell nuclear antigen (PCNA) and Ki-67 were used to assess liver regeneration. Results: At early phase (24 h after hepatectomy) it was demonstrated the negative effect of 5-fluorouracil on liver regeneration when assessed by Kwon formula (p < 0.0001), PCNA analysis (p = 0.02). With regeneration process complete (7 days), it was possible to demonstrate the sustained impairment of chemotherapy with 5-fluorouracil on hepatocytes regeneration phenomenon when measured by Kwon formula (p = 0.009), PCNA analysis (p = 0.0001) and Ki-67 analysis (0.001). Conclusions: Preoperative chemotherapy with intravenous 5-fluorouracil negatively affected the mechanisms of liver regeneration after major hepatectomy in rats.


Subject(s)
Animals , Rats , Chemoprevention/methods , Fluorouracil/therapeutic use , Hepatectomy/rehabilitation , Liver Regeneration/drug effects
18.
Cell Physiol Biochem ; 55(6): 739-760, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34816679

ABSTRACT

BACKGROUND/AIMS: Liver is considered as the vital organ in the body as it performs various essential functions. Following an injury to the liver, the repair process even though initially beneficial becomes pathogenic when it is not controlled appropriately. Extensive accumulation of extracellular matrix (ECM) components can ultimately lead to cirrhosis and liver failure. Thus, the ideal strategy to treat a liver injury is to generate new hepatocytes replacing damaged cells without causing excessive ECM deposition. The objective of this study was to evaluate the potential of mesenchymal stem cells, conditioned media and murine epidermal growth factor (m-EGF) in liver regeneration following partial hepatectomy in a rat model. METHODS: The animals were anaesthetized and a midline laparotomy was done. The liver was exposed and the left lateral and median lobes were ligated and resected out (about 65-70% of total liver mass). The muscles and skin were sutured in routine fashion and thus the rat model of partial hepatectomy was prepared. The animal models were equally distributed into 4 different groups namely A, B, C and D and treated with PBS, conditioned media, mesenchymal stem cells and epidermal growth factor respectively. The liver regeneration was assessed based on clinical, haemato-biochemical, colour imaging, histopathological and immune-histochemical parameters. RESULTS: Partial hepatectomy model with surgical removal of 65-70% liver lobe was standardized and successfully used in this study. Alkaline phosphatase (ALP), gamma glutamyl transferase (GGT), bilirubin, transaminases were significantly higher (P<0.05) in group A indicating that the liver damage was not restored properly. Colour digital imaging, histopathological and immune-histochemistry observations revealed that a better liver regeneration was observed in groups C and D, followed by groups B and A. Regeneration coefficient calculated based on liver weight was higher in groups C and D as compared to group A. CONCLUSION: Rat bone marrow-derived mesenchymal stem cells were found to induce hepatocytes proliferation; whereas EGF induced more angiogenesis. Conditioned media was not as effective as stem cells and EGF in liver tissue repair.


Subject(s)
Hepatectomy , Liver Regeneration/drug effects , Liver/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Culture Media, Conditioned/pharmacology , Female , Liver/surgery , Male , Rats , Rats, Wistar
19.
Life Sci ; 286: 120036, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34637793

ABSTRACT

AIMS: This study has been designed to investigate the role of vanillin either as prophylaxis or treatment in liver regeneration augmentation and liver fibrosis regression in thioacetamide (TAA) induced liver damage. MATERIALS AND METHODS: Animals were injected with TAA to induce liver injury (200mg/kg twice weekly) for 8 weeks. In vanillin prophylaxis group; rats were administered vanillin (100 mg/Kg; IP, daily) from day 1 of TAA injection for 8 weeks. In vanillin treatment group; rats were confronted with the same dose of TAA injection for 8 weeks then treated with vanillin (100 mg/Kg, IP, daily) for 4 weeks. ALT, AST activities, serum albumin, hepatic GSH, MDA, HGF, VEGF, IL-6 and TNF-α levels were measured and also, MMP-2, TIMP-1 and cyclin D gene expression were determined. Liver sections were stained with H&E and Sirius red and immunostained for Ki-67 and α-SMA for histological and immunohistological changes analysis. KEY FINDINGS: Vanillin improved liver function and histology. Also, showed a remarkable increase in hepatic HGF and VEGF level, and up-regulation of cyclin D1 expression accompanied by a significant up-regulation of MMP-2 and down- regulation of TIMP-1. All these effects were accompanied by TNF-α, IL-6 and oxidative stress significant attenuation. SIGNIFICANCE: In conclusion, vanillin enhanced liver regeneration in TAA induced liver damage model; targeting growth factors (HGF, VEGF) and cellular proliferation marker cyclin D1. As well as stimulating fibrosis regression by inhibition of ECM accumulation and enhancing its degradation.


Subject(s)
Benzaldehydes/pharmacology , Liver Cirrhosis/pathology , Liver Regeneration/drug effects , Animals , Benzaldehydes/metabolism , Cell Proliferation , Cyclin D1 , Intercellular Signaling Peptides and Proteins , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Regeneration/physiology , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Thioacetamide
20.
Exp Cell Res ; 409(1): 112866, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34655600

ABSTRACT

The occurrence of liver diseases is attributed to mitochondrial damage. Mitophagy selectively removes dysfunctional mitochondria, thereby preserving mitochondrial function. Augmenter of liver regeneration (ALR) protects the mitochondria from injury. However, whether ALR protection is associated with mitophagy remains unclear. In this study, mitochondrial damage was induced by carbonyl cyanide 3-chlorophenylhydrazone (CCCP), and long-form ALR (lfRNA)-mediated protection against this damage was investigated. Treatment of HepG2 cells with CCCP elevated the level of intracellular ROS, inhibited ATP production, and increased the mitochondrial membrane potential and cell apoptotic rate. However, in lfALR-transfected cells, CCCP-induced cell injury was clearly alleviated, the apoptosis and ROS levels clearly declined, and the ATP production was significantly enhanced as compared with that in vector-Tx cells. Furthermore, lfALR overexpression promoted autophagy and mitophagy via a PINK1/Parkin-dependent pathway, whereas knockdown of ALR suppressed mitophagy. In lfALR-transfected cells, the phosphorylation of AKT was decreased, thus, downregulating the phosphorylation of the transcription factor FOXO3a at Ser315. In contrast, the phosphorylation of AMPK was enhanced, thereby upregulating the phosphorylation of FOXO3a at Ser413. Consequently, FOXO3a's nuclear translocation and binding to the promoter region of PINK1 was enhanced, and the accumulation of PINK1/Parkin in mitochondria increased. Meanwhile, short-form ALR (sfALR) also increased PINK1 expression through FOXO3a with the similar pathway to lfALR. In conclusion, our data suggest a novel mechanism through which both lfALR and sfALR protect mitochondria by promoting PINK1/Parkin-dependent mitophagy through FOXO3a activation.


Subject(s)
Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Liver Regeneration/physiology , Mitochondria/metabolism , Mitophagy/physiology , Protein Kinases/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/drug effects , Autophagy/physiology , Cell Line, Tumor , Hep G2 Cells , Humans , Liver Regeneration/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitophagy/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL