Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Publication year range
1.
Cell ; 186(18): 3793-3809.e26, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37562401

ABSTRACT

Hepatocytes, the major metabolic hub of the body, execute functions that are human-specific, altered in human disease, and currently thought to be regulated through endocrine and cell-autonomous mechanisms. Here, we show that key metabolic functions of human hepatocytes are controlled by non-parenchymal cells (NPCs) in their microenvironment. We developed mice bearing human hepatic tissue composed of human hepatocytes and NPCs, including human immune, endothelial, and stellate cells. Humanized livers reproduce human liver architecture, perform vital human-specific metabolic/homeostatic processes, and model human pathologies, including fibrosis and non-alcoholic fatty liver disease (NAFLD). Leveraging species mismatch and lipidomics, we demonstrate that human NPCs control metabolic functions of human hepatocytes in a paracrine manner. Mechanistically, we uncover a species-specific interaction whereby WNT2 secreted by sinusoidal endothelial cells controls cholesterol uptake and bile acid conjugation in hepatocytes through receptor FZD5. These results reveal the essential microenvironmental regulation of hepatic metabolism and its human-specific aspects.


Subject(s)
Endothelial Cells , Liver , Animals , Humans , Mice , Endothelial Cells/metabolism , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver/cytology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Fibrosis/metabolism
2.
Mol Ther ; 32(4): 969-981, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38341614

ABSTRACT

The ability to target the native production site of factor VIII (FVIII)-liver sinusoidal endothelial cells (LSECs)-can improve the outcome of hemophilia A (HA) gene therapy. By testing a matrix of ultrasound-mediated gene delivery (UMGD) parameters for delivering a GFP plasmid into the livers of HA mice, we were able to define specific conditions for targeted gene delivery to different cell types in the liver. Subsequently, two conditions were selected for experiments to treat HA mice via UMGD of an endothelial-specific human FVIII plasmid: low energy (LE; 50 W/cm2, 150 µs pulse duration) to predominantly target endothelial cells or high energy (HE; 110 W/cm2, 150 µs pulse duration) to predominantly target hepatocytes. Both groups of UMGD-treated mice achieved persistent FVIII activity levels of ∼10% over 84 days post treatment; however, half of the HE-treated mice developed low-titer inhibitors while none of the LE mice did. Plasma transaminase levels and histological liver examinations revealed minimal transient liver damage that was lower in the LE group than in the HE group. These results indicate that UMGD can safely target LSECs with a lower-energy condition to achieve persistent FVIII gene expression, demonstrating that this novel technology is highly promising for therapeutic correction of HA.


Subject(s)
Factor VIII , Hemophilia A , Humans , Factor VIII/metabolism , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia A/pathology , Endothelial Cells/metabolism , Hepatocytes/metabolism , Liver/metabolism , Genetic Therapy/methods
3.
J Cell Physiol ; 239(5): e31198, 2024 May.
Article in English | MEDLINE | ID: mdl-38451745

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) dysfunction is a key process in the development of chronic liver disease (CLD). Progressive scarring increases liver stiffness in a winch-like loop stimulating a dysfunctional liver cell phenotype. Cellular stretching is supported by biomechanically modulated molecular factors (BMMFs) that can translocate into the cytoplasm to support mechanotransduction through cytoskeleton remodeling and gene transcription. Currently, the molecular mechanisms of stiffness-induced LSECs dysfunction remain largely unclear. Here we propose calcium- and integrin-binding protein 1 (CIB1) as BMMF with crucial role in LSECs mechanobiology in CLD. CIB1 expression and translocation was characterized in healthy and cirrhotic human livers and in LSECs cultured on polyacrylamide gels with healthy and cirrhotic-like stiffnesses. Following the modulation of CIB1 with siRNA, the transcriptome was scrutinized to understand downstream effects of CIB1 downregulation. CIB1 expression is increased in LSECs in human cirrhosis. In vitro, CIB1 emerges as an endothelial BMMF. In human umbilical vein endothelial cells and LSECs, CIB1 expression and localization are modulated by stiffness-induced trafficking across the nuclear membrane. LSECs from cirrhotic liver tissue both in animal model and human disease exhibit an increased amount of CIB1 in cytoplasm. Knockdown of CIB1 in LSECs exposed to high stiffness improves LSECs phenotype by regulating the intracellular tension as well as the inflammatory response. Our results demonstrate that CIB1 is a key factor in sustaining cellular tension and stretching in response to high stiffness. CIB1 downregulation ameliorates LSECs dysfunction, enhancing their redifferentiation, and reducing the inflammatory response.


Subject(s)
Calcium-Binding Proteins , Endothelial Cells , Liver Cirrhosis , Liver , Mechanotransduction, Cellular , Animals , Humans , Male , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cells, Cultured , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Female , Rats , Rats, Sprague-Dawley
4.
Curr Issues Mol Biol ; 46(8): 7997-8014, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39194690

ABSTRACT

Liver fibrosis is an important step in the transformation of chronic liver disease into cirrhosis and liver cancer, and structural changes and functional disorders of liver sinusoidal endothelial cells (LSECs) are early events in the occurrence of liver fibrosis. Therefore, it is necessary to identify the key regulatory genes of endothelial dysfunction in the process of liver fibrosis to provide a reference for the diagnosis and treatment of liver fibrosis. In this study, we identified 230 common differentially expressed genes (Co-DEGs) by analyzing transcriptomic data of primary LSECs from three different liver fibrosis mouse models (carbon tetrachloride; choline-deficient, l-amino acid-defined diet; and nonalcoholic steatohepatitis). Enrichment analysis revealed that the Co-DEGs were mainly involved in regulating the inflammatory response, immune response, angiogenesis, formation and degradation of the extracellular matrix, and mediating chemokine-related pathways. A Venn diagram analysis was used to identify 17 key genes related to the progression of liver cirrhosis. Regression analysis using the Lasso-Cox method identified genes related to prognosis among these key genes: SOX4, LGALS3, SERPINE2, CD52, and LPXN. In mouse models of liver fibrosis (bile duct ligation and carbon tetrachloride), all five key genes were upregulated in fibrotic livers. This study identified key regulatory genes for endothelial dysfunction in liver fibrosis, namely SOX4, LGALS3, SERPINE2, CD52, and LPXN, which will provide new targets for the development of therapeutic strategies targeting endothelial dysfunction in LSECs and liver fibrosis.

5.
J Hepatol ; 81(3): 543-561, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38763358

ABSTRACT

The hepatic sinusoids are composed of liver sinusoidal endothelial cells (LSECs), which are surrounded by hepatic stellate cells (HSCs) and contain liver-resident macrophages called Kupffer cells, and other patrolling immune cells. All these cells communicate with each other and with hepatocytes to maintain sinusoidal homeostasis and a spectrum of hepatic functions under healthy conditions. Sinusoidal homeostasis is disrupted by metabolites, toxins, viruses, and other pathological factors, leading to liver injury, chronic liver diseases, and cirrhosis. Alterations in hepatic sinusoids are linked to fibrosis progression and portal hypertension. LSECs are crucial regulators of cellular crosstalk within their microenvironment via angiocrine signaling. This review discusses the mechanisms by which angiocrine signaling orchestrates sinusoidal homeostasis, as well as the development of liver diseases. Here, we summarise the crosstalk between LSECs, HSCs, hepatocytes, cholangiocytes, and immune cells in health and disease and comment on potential novel therapeutic methods for treating liver diseases.


Subject(s)
Endothelial Cells , Hepatic Stellate Cells , Homeostasis , Liver Diseases , Signal Transduction , Humans , Homeostasis/physiology , Liver Diseases/metabolism , Liver Diseases/physiopathology , Signal Transduction/physiology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/physiology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Liver/metabolism , Liver/pathology , Animals , Hepatocytes/metabolism , Cell Communication/physiology , Kupffer Cells/physiology , Kupffer Cells/metabolism
6.
FASEB J ; 37(5): e22905, 2023 05.
Article in English | MEDLINE | ID: mdl-37039817

ABSTRACT

The hepatic vascular niche plays an important role in the pathological process of liver fibrosis. Liver sinusoidal endothelial cells (LSECs) predominantly compose hepatic vascular niches. Endothelial cell (EC)-expressing sphingosine 1-phosphate receptor 2 (S1pr2) plays an essential role in the regulation of vascular functions. Nevertheless, it remains unknown whether liver LSEC-S1pr2 might modulate pathological liver fibrosis. In this study, liver fibrosis was induced by hepatotoxin carbon tetrachloride (CCl4 ). The expression of S1pr2 is significantly downregulated in liver sinusoidal endothelial cells after CCl4 treatment. The loss of S1pr2 in LSECs significantly alleviated liver fibrosis after chronic insult, whereas the overexpression of S1pr2 in LSECs accentuated liver fibrogenesis. In vivo experiments further revealed that the deficiency of S1pr2 in LSECs dampened hepatic stellate cell (HSC) activation, while overexpression of S1pr2 in LSECs enhanced HSC activation with more extracellular matrix component production. Mechanistically, LSEC-S1pr2 activates the YAP signaling pathway to potentiate the transactivation of TGF-ß, which acts on HSCs in a paracrine manner, and thus aggravated liver fibrosis. Taken together, our results uncover a novel pathological mechanism of liver fibrosis in which LSEC-S1pr2 plays an important role in modulating the development of liver fibrosis, providing a future novel therapy target against liver fibrogenesis.


Subject(s)
Endothelial Cells , Liver Cirrhosis , Humans , Endothelial Cells/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Liver Cirrhosis/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
7.
Cell Commun Signal ; 22(1): 346, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943171

ABSTRACT

Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells that represent the interface between blood cells on one side and hepatocytes on the other side. LSECs not only form a barrier within the hepatic sinus, but also play important physiological functions such as regulating hepatic vascular pressure, anti-inflammatory and anti-fibrotic. Pathologically, pathogenic factors can induce LSECs capillarization, that is, loss of fenestra and dysfunction, which are conducive to early steatosis, lay the foundation for the progression of metabolic dysfunction-associated fatty liver disease (MAFLD), and accelerate metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis. The unique localization, phenotype, and function of LSECs make them potential candidates for reducing liver injury, inflammation, and preventing or reversing fibrosis in the future.


Subject(s)
Endothelial Cells , Liver , Humans , Endothelial Cells/metabolism , Endothelial Cells/pathology , Animals , Liver/metabolism , Liver/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology
8.
J Gastroenterol Hepatol ; 39(2): 224-230, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37939704

ABSTRACT

Liver ischemia/reperfusion injury (IRI) is a major complication after partial hepatectomy and liver transplantation and during hypovolemic shock and hypoxia-related diseases. Liver IRI is a current research hotspot. The early stage of liver IRI is characterized by injury and dysfunction of liver sinusoidal endothelial cells (LSECs), which, along with hepatocytes, are the major cells involved in liver injury. In this review, we elaborate on the roles played by LSECs in liver IRI, including the pathological features of LSECs, LSECs exacerbation of the sterile inflammatory response, LSECs interactions with platelets and the promotion of liver regeneration, and the activation of LSECs autophagy. In addition, we discuss the study of LSECs as therapeutic targets for the treatment of liver IRI and the existing problems when applying LSECs in liver IRI research.


Subject(s)
Endothelial Cells , Reperfusion Injury , Humans , Endothelial Cells/physiology , Liver/pathology , Hepatocytes/physiology , Reperfusion Injury/pathology , Ischemia/pathology
9.
J Gastroenterol Hepatol ; 39(7): 1413-1421, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38348885

ABSTRACT

BACKGROUND AND AIM: Safe radical hepatectomy is important for patients with colorectal liver metastases complicated by sinusoidal obstruction syndrome (SOS) after oxaliplatin-based chemotherapy. This study aimed to investigate the impact of preoperative administration of cilostazol (CZ), an oral selective phosphodiesterase III inhibitor, on hepatectomy in rat SOS model. MATERIAL AND METHODS: Rats were divided into NL (normal liver), SOS (monocrotaline [MCT]-treated), and SOS + CZ (MCT + CZ-treated) groups. MCT or CZ was administered orally, and a 30% partial hepatectomy was performed 48 h after MCT administration. Postoperative survival rates were evaluated (n = 9, for each). Other rats were sacrificed on postoperative days (POD) 1 and 3 and evaluated histologically, immunohistochemically, biochemically, and using transmission electron microscopy (TEM), focusing particularly on SOS findings, liver damage, and liver sinusoidal endothelial cell (LSEC) injury. RESULTS: The cumulative 10-day postoperative survival rate was significantly higher in the SOS + CZ group than in the SOS group (88.9% vs 33.3%, P = 0.001). Total SOS scores were significantly lower in the SOS + CZ group than in the SOS group on both POD 1 and 3. Serum biochemistry and immunohistochemistry showed that CZ reduced liver damage after hepatectomy. TEM revealed that LSECs were significantly preserved morphologically in the SOS + CZ group than in the SOS group on POD 1 (86.1 ± 8.2% vs 63.8 ± 9.3%, P = 0.003). CONCLUSION: Preoperative CZ administration reduced liver injury by protecting LSECs and improved the prognosis after hepatectomy in rats with SOS.


Subject(s)
Cilostazol , Disease Models, Animal , Hepatectomy , Hepatic Veno-Occlusive Disease , Phosphodiesterase 3 Inhibitors , Animals , Hepatic Veno-Occlusive Disease/prevention & control , Hepatic Veno-Occlusive Disease/etiology , Hepatic Veno-Occlusive Disease/pathology , Cilostazol/pharmacology , Hepatectomy/adverse effects , Male , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 3 Inhibitors/therapeutic use , Prognosis , Oxaliplatin/adverse effects , Liver Neoplasms/surgery , Liver Neoplasms/secondary , Liver Neoplasms/drug therapy , Survival Rate , Rats , Tetrazoles/administration & dosage , Tetrazoles/pharmacology , Colorectal Neoplasms/pathology , Liver/pathology , Rats, Sprague-Dawley
10.
Cell Biochem Funct ; 42(2): e3969, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38459746

ABSTRACT

The research of liver metastasis is a developing field. The ability of tumor cells to invade the liver depends on the complicated interactions between metastatic cells and local subpopulations in the liver (including Kupffer cells, hepatic stellate cells, liver sinusoidal endothelial cells, and immune-related cells). These interactions are mainly mediated by intercellular adhesion and the release of cytokines. Cell populations in the liver microenvironment can play a dual role in the progression of liver metastasis through different mechanisms. At the same time, we can see the participation of liver parenchymal cells and nonparenchymal cells in the process of liver metastasis of different tumors. Therefore, the purpose of this article is to summarize the relationship between cellular components of liver microenvironment and metastasis and emphasize the importance of different cells in the occurrence or potential regression of liver metastasis.


Subject(s)
Endothelial Cells , Liver Neoplasms , Humans , Endothelial Cells/pathology , Liver/pathology , Liver Neoplasms/pathology , Kupffer Cells , Hepatocytes , Tumor Microenvironment
11.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782474

ABSTRACT

Visualization of three-dimensional (3D) morphological changes in the subcellular structures of a biological specimen is a major challenge in life science. Here, we present an integrated chip-based optical nanoscopy combined with quantitative phase microscopy (QPM) to obtain 3D morphology of liver sinusoidal endothelial cells (LSEC). LSEC have unique morphology with small nanopores (50-300 nm in diameter) in the plasma membrane, called fenestrations. The fenestrations are grouped in discrete clusters, which are around 100 to 200 nm thick. Thus, imaging and quantification of fenestrations and sieve plate thickness require resolution and sensitivity of sub-100 nm along both the lateral and the axial directions, respectively. In chip-based nanoscopy, the optical waveguides are used both for hosting and illuminating the sample. The fluorescence signal is captured by an upright microscope, which is converted into a Linnik-type interferometer to sequentially acquire both superresolved images and phase information of the sample. The multimodal microscope provided an estimate of the fenestration diameter of 119 ± 53 nm and average thickness of the sieve plates of 136.6 ± 42.4 nm, assuming the constant refractive index of cell membrane to be 1.38. Further, LSEC were treated with cytochalasin B to demonstrate the possibility of precise detection in the cell height. The mean phase value of the fenestrated area in normal and treated cells was found to be 161 ± 50 mrad and 109 ± 49 mrad, respectively. The proposed multimodal technique offers nanoscale visualization of both the lateral size and the thickness map, which would be of broader interest in the fields of cell biology and bioimaging.


Subject(s)
Endothelial Cells/pathology , Endothelium/diagnostic imaging , Endothelium/pathology , Liver/diagnostic imaging , Microscopy/methods , Animals , Cell Membrane , Endothelium/metabolism , Fluorescence , Hepatocytes/pathology , Imaging, Three-Dimensional/methods , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy/instrumentation , Rats , Rats, Sprague-Dawley
12.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063116

ABSTRACT

The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Humans , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Animals , Myofibroblasts/metabolism , Myofibroblasts/pathology
13.
Biochem Biophys Res Commun ; 639: 20-28, 2023 01 08.
Article in English | MEDLINE | ID: mdl-36463757

ABSTRACT

Serum amyloid A (SAA) is an acute response protein that mainly produced by hepatocytes, and it can promote endothelial dysfunction via a pro-inflammatory and pro-thrombotic effect in atherosclerosis and renal disease. Overdose of Acetaminophen (APAP) will cause hepatotoxicity accompany with hepatocyte necrosis, liver sinusoidal endothelial cells (LSECs) damage and thrombosis in liver. However, whether SAA plays a role in APAP-induced liver toxicity remains unclear. Here, we evaluated the Saa1/2 expression in APAP-induced liver injury, and found that Saa1/2 production was significantly increased in an autocrine manner in APAP injury model. Moreover, we used neutralizing antibody (anti-SAA) to block the function of serum Saa1/2. We found that neutralizing serum Saa1/2 protected against APAP-induced liver injuries and increased the survival rate of mice that were treated with lethal dose APAP. Further investigations showed that blocking Saa1/2 reduced APAP-induced sinusoidal endothelium damage, hemorrhage and thrombosis. In addition, in vitro experiments showed that Saa1/2 augmented the toxic effect of APAP on LSECs, and Saa1/2 promoted platelets aggregation on LSECs cell membrane. Taken together, this study suggests that Saa1/2 may play a critical role in APAP-induced liver damages through platelets aggregation and sinusoidal damage. Therefore, we conceptually demonstrate that inhibition of SAA may be a potential intervention for APAP-directed acute liver injuries.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Mice , Animals , Acetaminophen/toxicity , Serum Amyloid A Protein/metabolism , Platelet Aggregation , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Endothelial Cells , Liver/metabolism , Hepatocytes/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism , Mice, Inbred C57BL
14.
Biochem Soc Trans ; 51(3): 1271-1277, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37264940

ABSTRACT

The liver is a highly organized organ that consists of hepatic parenchymal cells, hepatocytes, and non-parenchymal cells such as the liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), cholangiocytes, and Kupffer cells. Although previous studies have primarily focused on the hepatocyte dynamics in the injured liver, recent studies have shown that non-parenchymal cells play an essential role in both liver regeneration and liver fibrosis progression. Among the non-parenchymal cells, HSCs directly contribute to the progression of liver fibrosis because the activation of HSCs in response to liver injury or inflammation results in the excess production of extra cellular matrix. LSECs also contribute to modulate the function of hepatocytes, HSCs, and immune cells during liver fibrosis. Therefore, to investigate the mechanisms for liver fibrosis in vitro, it is necessary to develop an appropriate liver model that accurately recapitulates the pathology of human liver fibrosis including HSC activation. However, the supply of human cells is limited and freshly isolated liver cells easily lose their specific characteristics in culture. To overcome this shortage of human liver cells, human induced pluripotent stem cell (hiPSC)-derived liver cells were generated by mimicking the liver developmental process. In this review article, we outline the differentiation system of liver non-parenchymal cells from hiPSCs and development of in vitro liver disease models using hiPSC-derived liver cells. We describe the utility of these liver models as experimental systems to investigate the mechanism of liver fibrosis and development of drugs for the treatment thereof.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/pathology , Endothelial Cells , Hepatocytes , Liver Cirrhosis/therapy , Liver
15.
Hepatobiliary Pancreat Dis Int ; 22(1): 22-27, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36182636

ABSTRACT

Liver transplantation is the optimal treatment for patients with end-stage liver disease, metabolic liver diseases, and hepatic malignancies that are not amenable to resection. Hepatic ischemia-reperfusion injury (IRI) is the main problem in liver transplantation and liver resection, leading to parenchymal cell injury and organ dysfunction. The damage of liver sinusoidal endothelial cells (LSECs) is a critical event in IRI. LSECs work as an important regulating factor of liver regeneration after partial hepatectomy. This review primarily describes the mechanisms of LSECs injury in IRI and explores the roles of LSECs in liver regeneration, and briefly introduces the protective strategies targeting LSECs damaged in IRI.


Subject(s)
Liver Diseases , Reperfusion Injury , Humans , Endothelial Cells/metabolism , Endothelial Cells/pathology , Liver/pathology , Hepatocytes/pathology , Liver Diseases/pathology , Hepatectomy/adverse effects , Reperfusion Injury/etiology , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism
16.
Int J Mol Sci ; 24(15)2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37569282

ABSTRACT

Molecular markers of dedifferentiation of dysfunctional liver sinusoidal endothelial cells (LSEC) have not been fully elucidated. We aimed at deciphering the molecular profile of dysfunctional LSEC in different pathological scenarios. Flow cytometry was used to sort CD11b-/CD32b+ and CD11b-/CD32b- LSEC from three rat models of liver disease (bile duct ligation-BDL; inhaled carbon tetrachloride-CCl4; and high fat glucose/fructose diet-HFGFD). A full proteomic profile was performed applying nano-scale liquid chromatography tandem mass spectrometry (nLC-MS) and analyzed with PEAKS software. The percentage of CD32b- LSEC varied across groups, suggesting different capillarization processes. Both CD32+ and CD32b- LSEC from models are different from control LSEC, but differently expressed proteins in CD32b- LSEC are significantly higher. Heatmaps evidenced specific protein expression patterns for each model. Analysis of biological significance comparing dysfunctional CD32b- LSEC with specialized CD32b+ LSEC from controls showed central similarities represented by 45 common down-regulated proteins involved in the suppression of the endocytic machinery and 63 common up-regulated proteins associated with the actin-dependent cytoskeleton reorganization. In summary; substantial differences but also similarities in dysfunctional LSEC from the three most common models of liver disease were found, supporting the idea that LSEC may harbor different protein expression profiles according to the etiology or disease stage.


Subject(s)
Liver Diseases , Liver , Rats , Animals , Liver/metabolism , Endothelial Cells/metabolism , Proteomics , Liver Diseases/metabolism , Models, Theoretical
17.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047220

ABSTRACT

As the primary site for the biotransformation of drugs, the liver is the most focused on organ type in pharmaceutical research. However, despite being widely used in pharmaceutical research, animal models have inherent species differences, while two-dimensional (2D) liver cell monocultures or co-cultures and three-dimensional (3D) liver cell monoculture in vitro liver models do not sufficiently represent the complexity of the human liver's structure and function, making the evaluation results from these tools less reliable. Therefore, there is a pressing need to develop more representative in vitro liver models for pharmaceutical research. Fortunately, an exciting new development in recent years has been the emergence of 3D liver cell co-culture models. These models hold great promise as in vitro pharmaceutical research tools, because they can reproduce liver structure and function more practically. This review begins by explaining the structure and main cell composition of the liver, before introducing the potential advantages of 3D cell co-culture liver models for pharmaceutical research. We also discuss the main sources of hepatocytes and the 3D cell co-culture methods used in constructing these models. In addition, we explore the applications of 3D cell co-culture liver models with different functional states and suggest prospects for their further development.


Subject(s)
Pharmaceutical Research , Animals , Humans , Coculture Techniques , Liver , Hepatocytes/metabolism , Cell Culture Techniques/methods
18.
Zhonghua Gan Zang Bing Za Zhi ; 31(6): 668-672, 2023 Jun 20.
Article in Zh | MEDLINE | ID: mdl-37400397

ABSTRACT

Liver fibrosis incidence and adverse outcomes are high; however, there are no known chemical drugs or biological agents that are specific and effective for treatment. The paucity of a robust and realistic in vitro model for liver fibrosis is one of the major causes hindering anti-liver fibrosis drug development. This article summarizes the latest progress in the development of in vitro cell models for liver fibrosis, with a focus based on the analysis of induction and activation of hepatic stellate cells, cell co-culture, and 3D model co-construction, as well as concurrent potential methods based on hepatic sinusoidal endothelial cell establishment.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Humans , Liver Cirrhosis/pathology , Cell Culture Techniques , Endothelial Cells
19.
Zhonghua Gan Zang Bing Za Zhi ; 31(2): 212-215, 2023 Feb 20.
Article in Zh | MEDLINE | ID: mdl-37137841

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is widespread worldwide and thereby a very serious public health problem. There are currently no effective drug treatment measures. Liver sinusoidal endothelial cells (LSECs) are the most abundant non-parenchymal cells in the liver; however, it is still not clear what role LSECs play in NAFLD. This article reviews the research progress of LSECs in NAFLD in recent years in order to provide some reference for subsequent research.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Endothelial Cells , Liver , Hepatocytes
20.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(3): 469-474, 2023 May.
Article in Zh | MEDLINE | ID: mdl-37248570

ABSTRACT

Metastasis, a main cause of death in tumor patients, is a complicated process that involves multiple steps, presenting a major clinical challenge. Tumor cells break the physical boundaries of a primary tumor, intravasate into the lumina of blood vessels, travel around through blood circulation, extravasate into distant organs, colonize the host organs, and eventually develop into the foci of metastatic cancer. The metastasis of tumor cells exhibits organ-tropism, i.e., tumor cells preferentially spread to specific organs. Liver is a common site for metastasis. The pattern of metastasis in uveal melanoma, colorectal carcinoma, and pancreatic ductal adenocarcinoma shows organ-tropism for liver. The anatomical structure of liver determines its hemodynamic characteristics, e.g., low pressure and slow blood flow, which tend to facilitate the stasis and colonization of tumor cells in the liver. Besides the hemodynamic features, the metastatic colonization of liver depends largely on the interaction between tumor cells and the hepatic microenvironment (especially liver-resident cellular components). Resident cells of the hepatic microenvironment include hepatocytes, liver sinusoidal endothelial cells (LSECs), hepatic stellate cells (HSCs), Kupffer cells (KCs), etc. Herein, we discussed the role and significance of liver-resident cells in the metastatic colonization of tumor in the liver.


Subject(s)
Endothelial Cells , Liver Neoplasms , Humans , Liver/pathology , Hepatocytes , Kupffer Cells/pathology , Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Tumor Microenvironment/physiology
SELECTION OF CITATIONS
SEARCH DETAIL