Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.112
Filter
2.
Expert Opin Ther Pat ; 34(3): 127-140, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38557201

ABSTRACT

INTRODUCTION: HMGB1 is a non-histone chromatin protein released or secreted in response to tissue damage or infection. Extracellular HMGB1, as a crucial immunomodulatory factor, binds with several different receptors to innate inflammatory responses that aggravate acute and chronic liver diseases. The increased levels of HMGB1 have been reported in various liver diseases, highlighting that it represents a potential biomarker and druggable target for therapeutic development. AREAS COVERED: This review summarizes the current knowledge on the structure, function, and interacting receptors of HMGB1 and its significance in multiple liver diseases. The latest patented and preclinical studies of HMGB1 inhibitors (antibodies, peptides, and small molecules) for liver diseases are summarized by using the keywords 'HMGB1,' 'HMGB1 antagonist, HMGB1-inhibitor,' 'liver disease' in Web of Science, Google Scholar, Google Patents, and PubMed databases in the year from 2017 to 2023. EXPERT OPINIONS: In recent years, extensive research on HMGB1-dependent inflammatory signaling has discovered potent inhibitors of HMGB1 to reduce the severity of liver injury. Despite significant progress in the development of HMGB1 antagonists, few of them are approved for clinical treatment of liver-related diseases. Developing safe and effective specific inhibitors for different HMGB1 isoforms and their interaction with receptors is the focus of future research.


Subject(s)
Drug Development , HMGB1 Protein , Liver Diseases , Patents as Topic , Humans , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/metabolism , Liver Diseases/drug therapy , Liver Diseases/physiopathology , Animals , Biomarkers/metabolism , Inflammation/drug therapy , Inflammation/physiopathology , Severity of Illness Index
3.
Auton Neurosci ; 253: 103174, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38579493

ABSTRACT

The liver is a large organ with crucial functions in metabolism and immune defense, as well as blood homeostasis and detoxification, and it is clearly in bidirectional communication with the brain and rest of the body via both neural and humoral pathways. A host of neural sensory mechanisms have been proposed, but in contrast to the gut-brain axis, details for both the exact site and molecular signaling steps of their peripheral transduction mechanisms are generally lacking. Similarly, knowledge about function-specific sensory and motor components of both vagal and spinal access pathways to the hepatic parenchyma is missing. Lack of progress largely owes to controversies regarding selectivity of vagal access pathways and extent of hepatocyte innervation. In contrast, there is considerable evidence for glucose sensors in the wall of the hepatic portal vein and their importance for glucose handling by the liver and the brain and the systemic response to hypoglycemia. As liver diseases are on the rise globally, and there are intriguing associations between liver diseases and mental illnesses, it will be important to further dissect and identify both neural and humoral pathways that mediate hepatocyte-specific signals to relevant brain areas. The question of whether and how sensations from the liver contribute to interoceptive self-awareness has not yet been explored.


Subject(s)
Interoception , Liver Diseases , Liver , Humans , Interoception/physiology , Animals , Liver Diseases/physiopathology , Liver Diseases/metabolism , Liver/metabolism , Brain/metabolism , Brain/physiology
4.
J Heart Lung Transplant ; 43(7): 1105-1115, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38373557

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) can lead to congestive hepatopathy, known as cardiohepatic syndrome (CHS). Hepatic congestion is associated with increased liver stiffness, which can be quantified using shear wave elastography. We aimed to investigate whether hepatic shear wave elastography detects patients at risk in the early stages of PH. METHODS: Sixty-three prospectively enrolled patients undergoing right heart catheterization (52 diagnosed with PH and 11 with invasive exclusion of PH) and 52 healthy volunteers underwent assessments including echocardiography and hepatic shear wave elastography. CHS was defined as increased levels of ≥2 of the following: gamma-glutamyl transferase, alkaline phosphatase, and bilirubin. Liver stiffness was defined as normal (≤5.0 kPa) or high (>5.0 kPa). RESULTS: Compared with normal liver stiffness, high liver stiffness was associated with impaired right ventricular (RV) and right atrial (RA) function (median [interquartile range] RV ejection fraction: 54 [49; 57]% vs 45 [34; 51]%, p < 0.001; RA reservoir strain: 49 [41; 54]% vs 33 [22; 41]%, p < 0.001), more severe tricuspid insufficiency (p < 0.001), and higher prevalence of hepatovenous backflow (2% vs 29%, p < 0.001) and CHS (2% vs 10%, p = 0.038). In the patient subgroup with precapillary PH (n = 48), CHS and high liver stiffness were associated with increased European Society of Cardiology/European Respiratory Society 2022 risk scores (p = 0.003). CONCLUSIONS: Shear wave liver elastography yields important information regarding right heart function and may complement risk assessment in patients with (suspected) PH.


Subject(s)
Elasticity Imaging Techniques , Hypertension, Pulmonary , Liver , Ventricular Dysfunction, Right , Humans , Female , Male , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/etiology , Middle Aged , Elasticity Imaging Techniques/methods , Prospective Studies , Prognosis , Ventricular Dysfunction, Right/physiopathology , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/diagnosis , Liver/diagnostic imaging , Liver/physiopathology , Syndrome , Cardiac Catheterization , Adult , Liver Diseases/physiopathology , Liver Diseases/complications , Echocardiography
6.
Hepatology ; 77(2): 619-639, 2023 02 01.
Article in English | MEDLINE | ID: mdl-35524448

ABSTRACT

The endoplasmic reticulum (ER) is an intracellular organelle that fosters the correct folding of linear polypeptides and proteins, a process tightly governed by the ER-resident enzymes and chaperones. Failure to shape the proper 3-dimensional architecture of proteins culminates in the accumulation of misfolded or unfolded proteins within the ER, disturbs ER homeostasis, and leads to canonically defined ER stress. Recent studies have elucidated that cellular perturbations, such as lipotoxicity, can also lead to ER stress. In response to ER stress, the unfolded protein response (UPR) is activated to reestablish ER homeostasis ("adaptive UPR"), or, conversely, to provoke cell death when ER stress is overwhelmed and sustained ("maladaptive UPR"). It is well documented that ER stress contributes to the onset and progression of multiple hepatic pathologies including NAFLD, alcohol-associated liver disease, viral hepatitis, liver ischemia, drug toxicity, and liver cancers. Here, we review key studies dealing with the emerging role of ER stress and the UPR in the pathophysiology of liver diseases from cellular, murine, and human models. Specifically, we will summarize current available knowledge on pharmacological and non-pharmacological interventions that may be used to target maladaptive UPR for the treatment of nonmalignant liver diseases.


Subject(s)
Endoplasmic Reticulum Stress , Liver Diseases , Animals , Humans , Mice , Endoplasmic Reticulum Stress/physiology , Liver Diseases, Alcoholic , Molecular Chaperones , Non-alcoholic Fatty Liver Disease , Unfolded Protein Response , Liver Diseases/physiopathology
7.
PeerJ ; 10: e14299, 2022.
Article in English | MEDLINE | ID: mdl-36523459

ABSTRACT

Liver fibrosis develops as a result of severe liver damage and is considered a major clinical concern throughout the world. Many factors are crucial for liver fibrosis progression. While advancements have been made to understand this disease, no effective pharmacological drug and treatment strategies have been established that can effectively prevent liver fibrosis or even could halt the fibrotic process. Most of those advances in curing liver fibrosis have been aimed towards mitigating the causes of fibrosis, including the development of potent antivirals to inhibit the hepatitis virus. It is not practicable for many individuals; however, a liver transplant becomes the only suitable alternative. A liver transplant is an expensive procedure. Thus, there is a significant need to identify potential targets of liver fibrosis and the development of such agents that can effectively treat or reverse liver fibrosis by targeting them. Researchers have identified hypoxia-inducible factors (HIFs) in the last 16 years as important transcription factors driving several facets of liver fibrosis, making them possible therapeutic targets. The latest knowledge on HIFs and their possible role in liver fibrosis, along with the cell-specific activities of such transcription factors that how they play role in liver fibrosis progression, is discussed in this review.


Subject(s)
Liver Cirrhosis , Transcription Factors , Humans , Cell Hypoxia , Hypoxia , Liver Cirrhosis/drug therapy , Liver Cirrhosis/physiopathology , Liver Diseases/etiology , Liver Diseases/physiopathology , Transcription Factors/physiology , Transcription Factors/therapeutic use
8.
Int J Mol Sci ; 23(17)2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36077452

ABSTRACT

Metabolic associated fatty liver disease (MAFLD) is one of the most common causes of chronic liver disease worldwide. To date, there is no FDA-approved treatment, so there is an urgent need to determine its pathophysiology and underlying molecular mechanisms. Autophagy is a lysosomal degradation pathway that removes damaged organelles and misfolded proteins after cell injury through endoplasmic reticulum stress or starvation, which inhibits apoptosis and promotes cell survival. Recent studies have shown that autophagy plays an important role in removing lipid droplets from hepatocytes. Autophagy has also been reported to inhibit the production of pro-inflammatory cytokines and provide energy for the hepatic stellate cells activation during liver fibrosis. Thyroid hormone, irisin, melatonin, hydrogen sulfide, sulforaphane, DA-1241, vacuole membrane protein 1, nuclear factor erythroid 2-related factor 2, sodium-glucose co-transporter type-2 inhibitors, immunity-related GTPase M, and autophagy-related gene 7 have been reported to ameliorate MAFLD via autophagic induction. Lipid receptor CD36, SARS-CoV-2 Spike protein and leucine aminopeptidase 3 play a negative role in the autophagic function. This review summarizes recent advances in the role of autophagy in MAFLD. Autophagy modulates major pathological changes, including hepatic lipid metabolism, inflammation, and fibrosis, suggesting the potential of modulating autophagy for the treatment of MAFLD.


Subject(s)
Autophagy , Liver Diseases , Non-alcoholic Fatty Liver Disease , Humans , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/physiopathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology
9.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216473

ABSTRACT

Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.


Subject(s)
Apoptosis , Liver Diseases/physiopathology , Mitochondria/physiology , Reperfusion Injury/physiopathology , Animals , Endoplasmic Reticulum Stress , Humans
10.
Gut Microbes ; 14(1): 2027853, 2022.
Article in English | MEDLINE | ID: mdl-35129072

ABSTRACT

The intestinal flora plays an important role in the development of many human and animal diseases. Microbiome association studies revealed the potential regulatory function of intestinal bacteria in many liver diseases, such as autoimmune hepatitis, viral hepatitis and alcoholic hepatitis. However, the key intestinal bacterial strains that affect pathological liver injury and the underlying functional mechanisms remain unclear. We found that the gut microbiota from gentamycin (Gen)-treated mice significantly alleviated concanavalin A (ConA)-induced liver injury compared to vancomycin (Van)-treated mice by inhibiting CD95 expression on the surface of hepatocytes and reducing CD95/CD95L-mediated hepatocyte apoptosis. Through the combination of microbiota sequencing and correlation analysis, we isolated 5 strains with the highest relative abundance, Bacteroides acidifaciens (BA), Parabacteroides distasonis (PD), Bacteroides thetaiotaomicron (BT), Bacteroides dorei (BD) and Bacteroides uniformis (BU), from the feces of Gen-treated mice. Only BA played a protective role against ConA-induced liver injury. Further studies demonstrated that BA-reconstituted mice had reduced CD95/CD95L signaling, which was required for the decrease in the L-glutathione/glutathione (GSSG/GSH) ratio observed in the liver. BA-reconstituted mice were also more resistant to alcoholic liver injury. Our work showed that a specific murine intestinal bacterial strain, BA, ameliorated liver injury by reducing hepatocyte apoptosis in a CD95-dependent manner. Determination of the function of BA may provide an opportunity for its future use as a treatment for liver disease.


Subject(s)
Bacteroides/physiology , Gastrointestinal Microbiome , Liver Diseases/prevention & control , fas Receptor/metabolism , Animals , Apoptosis , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteroides/genetics , Bacteroides/isolation & purification , Feces/microbiology , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver Diseases/metabolism , Liver Diseases/microbiology , Liver Diseases/physiopathology , Mice , Mice, Inbred C57BL , fas Receptor/genetics
11.
BMC Cardiovasc Disord ; 22(1): 51, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35164689

ABSTRACT

BACKGROUND: To investigate whether liver stiffness (LS) can predict adverse cardiac events in Chinese patients with heart failure (HF). METHODS: A total of 53 hospitalized patients with HF were enrolled, and LS and tricuspid annual plane systolic excursion (TAPSE) were determined with Fibroscan® and echocardiography before discharge. They were divided into two groups: high LS group (LS > 6.9 Kpa, n = 23) and low LS group (LS ≤ 6.9 Kpa, n = 30). Patients were followed up for 24 months at an interval of 3 months. The endpoint of follow-up was death or rehospitalization for HF. RESULTS: All patients were followed up for 24 months or until the endpoint. Patients in the high LS group had lower platelet count (P = 0.014), lower creatine clear rate (P = 0.014), higher level of B-type natriuretic peptide at discharge (P = 0.012), and lower TAPSE (P < 0.001) than those in the low LS group. During 24 months of follow-up, 3 (5.7%) deaths and 21 (39.6%) hospitalizations for HF were observed. Patients in the high LS group had a higher rate of death/rehospitalization than those in the low LS group (Hazard ratio 4.81; 95% confidence interval 1.69-13.7, P = 0.003) after adjustment for age, sex, platelet count, creatine clear rate, and B-type natriuretic peptide level. Moreover, TAPSE ≤ 16 could predict adverse cardiac events with an HR of 6.63 (95% confidence interval 1.69-13.7, P = 0.004) after adjustment for age, sex, platelet count, creatine clear rate, and B-type natriuretic peptide level. CONCLUSION: LS and TAPSE could be used to predict worse outcomes in patients with HF.


Subject(s)
Echocardiography , Elasticity Imaging Techniques , Heart Failure/diagnostic imaging , Liver Diseases/diagnostic imaging , Aged , China , Disease Progression , Female , Heart Failure/mortality , Heart Failure/physiopathology , Heart Failure/therapy , Hospitalization , Humans , Liver Diseases/mortality , Liver Diseases/physiopathology , Male , Middle Aged , Patient Readmission , Predictive Value of Tests , Prognosis , Prospective Studies , Risk Assessment , Risk Factors , Time Factors
12.
Cells ; 11(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-35053347

ABSTRACT

Changes in the structure and function of blood vessels are important factors that play a primary role in regeneration of injured organs. WKYMVm has been reported as a therapeutic factor that promotes the migration and proliferation of angiogenic cells. Additionally, we previously demonstrated that placenta-derived mesenchymal stem cells (PD-MSCs) induce hepatic regeneration in hepatic failure via antifibrotic effects. Therefore, our objectives were to analyze the combination effect of PD-MSCs and WKYMVm in a rat model with bile duct ligation (BDL) and evaluate their therapeutic mechanism. To analyze the anti-fibrotic and angiogenic effects on liver regeneration, it was analyzed using ELISA, qRT-PCR, Western blot, immunofluorescence, and immunohistochemistry. Collagen accumulation was significantly decreased in PD-MSCs with the WKYMVm combination (Tx+WK) group compared with the nontransplantation (NTx) and PD-MSC-transplanted (Tx) group (p < 0.05). Furthermore, the combination of PD-MSCs with WKYMVm significantly promoted hepatic function by increasing hepatocyte proliferation and albumin as well as angiogenesis by activated FPR2 signaling (p < 0.05). The combination therapy of PD-MSCs with WKYMVm could be an efficient treatment in hepatic diseases via vascular remodeling. Therefore, the combination therapy of PD-MSCs with WKYMVm could be a new therapeutic strategy in degenerative medicine.


Subject(s)
Liver Diseases/physiopathology , Liver Diseases/therapy , Liver/physiopathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Oligopeptides/pharmacology , Placenta/cytology , Vascular Remodeling , Animals , Combined Modality Therapy , Disease Models, Animal , Female , Liver/drug effects , Pregnancy , Rats , Vascular Remodeling/drug effects
14.
Food Chem Toxicol ; 160: 112814, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34999178

ABSTRACT

We investigated whether diallyl disulfide (DADS) has protective effects against 1,3-dichloro-2-propanol (1,3-DCP)-induced hepatotoxicity and oxidative damage in rats and HepG2 cells. DADS was administered to rats once daily for 7 days at doses of 30 and 60 mg/kg/day. One hour after the final DADS treatment, the rats were administered 90 mg/kg 1,3-DCP to induce acute hepatotoxicity. DADS treatment significantly suppressed the increase in serum aminotransferase levels induced by 1,3-DCP administration, and reduced histopathological alterations in the liver. DADS treatment reduced 1-3-DCP-induced apoptotic changes in the liver, as revealed by terminal deoxynucleotidyl transferase dUTP nick end labeling staining and immunohistochemistry for caspase-3. DADS treatment competitively inhibited or reduced cytochrome p450 2E1 (CYP2E1) expression, which is involved in the metabolic activation of 1,3-DCP, and enhanced antioxidant properties. Furthermore, DADS treatment inhibited phosphorylation of mitogen-activated protein kinases (MAPKs) and apoptotic signaling. In in vitro experiments, MAPKs inhibitors reduced the expression of Bax/Bcl-2/Caspase 3 signaling, which effects were more significant in co-treated cells with DADS and MAPKs inhibitors. In conclusion, the protective effect of DADS against 1,3-DCP-induced hepatotoxicity may be related to blocking the metabolic activation of 1,3-DCP by suppressing CYP2E1 expression, inducing antioxidant enzyme activity, and reducing apoptotic activity by inhibiting phosphorylation of MAPKs.


Subject(s)
Allyl Compounds/administration & dosage , Disulfides/administration & dosage , Liver Diseases/prevention & control , Mitogen-Activated Protein Kinases/metabolism , Protective Agents/pharmacology , alpha-Chlorohydrin/analogs & derivatives , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Hep G2 Cells , Humans , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/physiopathology , Male , Mitogen-Activated Protein Kinases/genetics , Oxidative Stress/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , alpha-Chlorohydrin/toxicity
15.
Pediatrics ; 149(1 Suppl 1): S59-S65, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34970684

ABSTRACT

CONTEXT: Develop evidence-based criteria for individual organ dysfunction. OBJECTIVES: Evaluate current evidence and develop contemporary consensus criteria for acute liver dysfunction with associated outcomes in critically ill children. DATA SOURCES: Electronic searches of PubMed and Embase conducted from January 1992 to January 2020, used medical subject heading terms and text words to characterize acute liver dysfunction and outcomes. STUDY SELECTION: Studies evaluating critically ill children with acute liver dysfunction, assessed screening tools, and outcomes were included. Studies evaluating adults, infants ≤36 weeks gestational age, or animals or were reviews/commentaries, case series with sample size ≤10, or non-English language studies were excluded. DATA EXTRACTION: Data were abstracted from each eligible study into a data extraction form along with risk of bias assessment by a task force member. RESULTS: The systematic review supports criteria for acute liver dysfunction, in the absence of known chronic liver disease, as having onset of symptoms <8 weeks, combined with biochemical evidence of acute liver injury, and liver-based coagulopathy, with hepatic encephalopathy required for an international normalized ratio between 1.5 and 2.0. LIMITATIONS: Unable to assess acute-on-chronic liver dysfunction, subjective nature of hepatic encephalopathy, relevant articles missed by reviewers. CONCLUSIONS: Proposed criteria identify an infant, child, or adolescent who has reached a clinical threshold where any of the 3 outcomes (alive with native liver, death, or liver transplant) are possible and should prompt an urgent liaison with a recognized pediatric liver transplant center if liver failure is the principal driver of multiple organ dysfunction.


Subject(s)
Liver Diseases/diagnosis , Multiple Organ Failure/diagnosis , Adolescent , Child , Critical Illness , Humans , Infant , Liver Diseases/physiopathology , Multiple Organ Failure/physiopathology , Organ Dysfunction Scores
16.
Basic Clin Pharmacol Toxicol ; 130 Suppl 1: 5-15, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33872466

ABSTRACT

Drug development is a failure-prone endeavour, and more than 85% of drugs fail during clinical development, showcasing that current preclinical systems for compound selection are clearly inadequate. Liver toxicity remains a major reason for safety failures. Furthermore, all efforts to develop pharmacological therapies for a variety of chronic liver diseases, such as non-alcoholic steatohepatitis (NASH) and fibrosis, remain unsuccessful. Considering the time and expense of clinical trials, as well as the substantial burden on patients, new strategies are thus of paramount importance to increase clinical success rates. To this end, human liver spheroids are becoming increasingly utilized as they allow to preserve patient-specific phenotypes and functions for multiple weeks in culture. We here review the recent application of such systems for i) predictive and mechanistic analyses of drug hepatotoxicity, ii) the evaluation of hepatic disposition and metabolite formation of low clearance drugs and iii) the development of drugs for metabolic and infectious liver diseases, including NASH, fibrosis, malaria and viral hepatitis. We envision that with increasing dissemination, liver spheroids might become the new gold standard for such applications in translational pharmacology and toxicology.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Liver Diseases/drug therapy , Spheroids, Cellular/drug effects , Animals , Chemical and Drug Induced Liver Injury/prevention & control , Drug Development/methods , Humans , Liver/drug effects , Liver/pathology , Liver Diseases/physiopathology , Pharmacology/methods , Spheroids, Cellular/pathology , Toxicology/methods , Translational Research, Biomedical/methods
17.
J Hepatol ; 76(2): 420-434, 2022 02.
Article in English | MEDLINE | ID: mdl-34678405

ABSTRACT

Cystic fibrosis (CF) is the most common autosomal recessive disease in the Caucasian population. Cystic fibrosis-related liver disease (CFLD) is defined as the pathogenesis related to the underlying CFTR defect in biliary epithelial cells. CFLD needs to be distinguished from other liver manifestations that may not have any pathological significance. The clinical/histological presentation and severity of CFLD vary. The main histological presentation of CFLD is focal biliary fibrosis, which is usually asymptomatic. Portal hypertension develops in a minority of cases (about 10%) and may require specific management including liver transplantation for end-stage liver disease. Portal hypertension is usually the result of the progression of focal biliary fibrosis to multilobular cirrhosis during childhood. Nevertheless, non-cirrhotic portal hypertension as a result of porto-sinusoidal vascular disease is now identified increasingly more frequently, mainly in young adults. To evaluate the effect of new CFTR modulator therapies on the liver, the spectrum of hepatobiliary involvement must first be precisely classified. This paper discusses the phenotypic features of CFLD, its underlying physiopathology and relevant diagnostic and follow-up approaches, with a special focus on imaging.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/complications , Liver Diseases/etiology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/therapeutic use , Elasticity Imaging Techniques/methods , Elasticity Imaging Techniques/statistics & numerical data , Humans , Hypertension, Portal/diagnostic imaging , Hypertension, Portal/physiopathology , Liver/pathology , Liver Diseases/diagnostic imaging , Liver Diseases/physiopathology , Severity of Illness Index , Ultrasonography/methods , Ultrasonography/statistics & numerical data
18.
J Sci Food Agric ; 102(3): 1066-1075, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34309869

ABSTRACT

BACKGROUND: Inflammation-related diseases present a significant public health problem. Ginger is a flavoring spice and medicinal herb with anti-inflammatory activity. This study investigated the preventive effects of ginger extract (GE) and its main bioactive component, 6-gingerol (6G), on lipopolysaccharide (LPS)-induced intestinal barrier dysfunction and liver injury in mice. RESULTS: GE and 6G were orally administered to mice for seven consecutive days before LPS administration. After 24 h, the mice were sacrificed. GE and 6G were found to significantly reverse LPS-induced inflammation in the mouse ileum by modifying the NF-κB pathway. They also alleviated apoptosis in the ileum by downregulating Bax and cytochrome c gene expression and by inhibiting the caspase-3 pathway. Through the aforementioned mechanisms, GE and 6G restored the intestinal barrier by increasing ZO-1 and claudin-1 protein expressions. Gut-derived LPS induced inflammation and apoptosis in the liver; these effects were markedly reversed through GE and 6G treatment. 6G was the most abundant component in GE, as evidenced through liquid chromatography-mass spectrometry, and accounted for >50% of total gingerols and shogaols in GE. CONCLUSION: The current results support the use of GE and 6G as dietary supplements to protect against gut-derived endotoxemia-associated inflammatory response and disorders. © 2021 Society of Chemical Industry.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Catechols/administration & dosage , Fatty Alcohols/administration & dosage , Intestinal Diseases/drug therapy , Liver Diseases/drug therapy , Plant Extracts/administration & dosage , Zingiber officinale/chemistry , Animals , Apoptosis/drug effects , Humans , Intestinal Diseases/immunology , Intestinal Diseases/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/injuries , Lipopolysaccharides/adverse effects , Liver/drug effects , Liver/immunology , Liver/injuries , Liver Diseases/immunology , Liver Diseases/physiopathology , Male , Mice , Mice, Inbred ICR
19.
Hepatol Commun ; 6(2): 255-269, 2022 02.
Article in English | MEDLINE | ID: mdl-34658172

ABSTRACT

Liver injury, characterized predominantly by elevated aspartate aminotransferase and alanine aminotransferase, is a common feature of coronavirus disease 2019 (COVID-19) symptoms caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). Additionally, SARS-CoV-2 infection is associated with acute-on-chronic liver failure in patients with cirrhosis and has a notably elevated mortality in patients with alcohol-related liver disease compared to other etiologies. Direct viral infection of the liver with SARS-CoV-2 remains controversial, and alternative pathophysiologic explanations for its hepatic effects are an area of active investigation. In this review, we discuss the effects of SARS-CoV-2 and the inflammatory environment it creates on endothelial cells and platelets more generally and then with a hepatic focus. In doing this, we present vascular inflammation and thrombosis as a potential mechanism of liver injury and liver-related complications in COVID-19.


Subject(s)
Blood Platelet Disorders/virology , COVID-19/physiopathology , Endothelium, Vascular/virology , Inflammation/virology , Liver Diseases/virology , Thrombosis/virology , Blood Platelet Disorders/immunology , Blood Platelet Disorders/physiopathology , COVID-19/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Humans , Inflammation/immunology , Inflammation/physiopathology , Liver Diseases/immunology , Liver Diseases/physiopathology , Thrombosis/immunology , Thrombosis/physiopathology
20.
Hepatology ; 75(2): 379-390, 2022 02.
Article in English | MEDLINE | ID: mdl-34510513

ABSTRACT

BACKGROUND AND AIMS: Although magnetic resonance elastography (MRE) has been well-established for detecting and staging liver fibrosis, its prognostic role in determining outcomes of chronic liver disease (CLD) is mostly unknown. APPROACH AND RESULTS: This retrospective study consisted of 1269 subjects who underwent MRE between 2007 and 2009 and followed up until death or last known clinical encounter or end of study period. Charts were reviewed for cirrhosis development, decompensation, and transplant or death. The cohort was split into baseline noncirrhosis (group 1), compensated cirrhosis (group 2), and decompensated cirrhosis (group 3). Cox-regression analysis with age, sex, splenomegaly, CLD etiology, Child-Pugh Score (CPS), Fibrosis-4 Index (FIB-4) score, and Model for End-Stage Liver Disease (MELD)-adjusted HR for every 1-kPa increase in liver stiffness measurement (LSM) were used to assess the predictive performance of MRE on outcomes. Group 1 (n = 821) had baseline median LSM of 2.8 kPa, and cirrhosis developed in 72 (8.8%) subjects with an overall rate of about 1% cirrhosis/year. Baseline LSM predicted the future cirrhosis with multivariable adjusted HR of 2.38 (p < 0.0001) (concordance, 0.84). In group 2 (n = 277) with baseline median LSM of 5.7 kPa, 83 (30%) subjects developed decompensation. Baseline LSM predicted the future decompensation in cirrhosis with FIB-4 and MELD-adjusted HR of 1.22 (p < 0.0001) (concordance, 0.75). In group 3 (n = 171) with median baseline LSM of 6.8 kPa (5.2, 8.4), 113 (66%) subjects had either death or transplant. Baseline LSM predicted the future transplant or death with HR of 1.11 (p = 0.013) (concordance 0.53) but not in CPS and MELD-adjusted models (p = 0.08). CONCLUSION: MRE-based LSM is independently predictive of development of future cirrhosis and decompensation, and has predictive value in future transplant/death in patients with CLD.


Subject(s)
Elasticity Imaging Techniques , Liver Diseases/diagnostic imaging , Liver Diseases/physiopathology , Adult , Aged , Carcinoma, Hepatocellular/etiology , Chronic Disease , Disease Progression , Elasticity , Female , Humans , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/etiology , Liver Cirrhosis/physiopathology , Liver Cirrhosis/surgery , Liver Diseases/complications , Liver Neoplasms/etiology , Liver Transplantation , Longitudinal Studies , Male , Middle Aged , Predictive Value of Tests , Prognosis , Retrospective Studies , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...