Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Hepatol Commun ; 8(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38780301

ABSTRACT

BACKGROUND: Vertical sleeve gastrectomy (SGx) is a type of bariatric surgery to treat morbid obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). The molecular mechanisms of SGx to improve MASLD are unclear, but increased bile acids (BAs) and FGF19 (mouse FGF15) were observed. FGF15/19 is expressed in the ileum in response to BAs and is critical in not only suppressing BA synthesis in the liver but also promoting energy expenditure. We hypothesized the reduction of obesity and resolution of MASLD by SGx may be mediated by FGF15/19. METHODS: First, we conducted hepatic gene expression analysis in obese patients undergoing SGx, with the results showing increased expression of FGF19 in obese patients' livers. Next, we used wild-type and intestine-specific Fgf15 knockout mice (Fgf15ile-/-) to determine the effects of FGF15 deficiency on improving the metabolic effects. RESULTS: SGx improved metabolic endpoints in both genotypes, evidenced by decreased obesity, improved glucose tolerance, and reduced MASLD progression. However, Fgf15ile-/- mice showed better improvement compared to wild-type mice after SGx, suggesting that other mediators than FGF15 are also responsible for the beneficial effects of FGF15 deficiency. Further gene expression analysis in brown adipose tissue suggests increased thermogenesis. CONCLUSIONS: FGF15 deficiency, the larger BA pool and higher levels of secondary BAs may increase energy expenditure in extrahepatic tissues, which may be responsible for improved metabolic functions following SGx.


Subject(s)
Fatty Liver , Fibroblast Growth Factors , Gastrectomy , Mice, Knockout , Obesity, Morbid , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Animals , Gastrectomy/methods , Mice , Obesity, Morbid/surgery , Obesity, Morbid/genetics , Obesity, Morbid/metabolism , Humans , Male , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Bile Acids and Salts/metabolism , Liver/metabolism , Adult , Middle Aged , Bariatric Surgery , Mice, Inbred C57BL
2.
Noncoding RNA ; 10(2)2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38668386

ABSTRACT

The long non-coding RNA (lncRNA) hepatocyte nuclear factor-1 alpha (HNF1A) antisense RNA 1 (HNF1A-AS1) is an important lncRNA for liver growth, development, cell differentiation, and drug metabolism. Like many lncRNAs, HNF1A-AS1 has multiple annotated alternative transcripts in the human genome. Several fundamental biological questions are still not solved: (1) How many transcripts really exist in biological samples, such as liver samples and liver cell lines? (2) What are the expression patterns of different alternative HNF1A-AS1 transcripts at different conditions, including during cell growth and development, after exposure to xenobiotics (such as drugs), and in disease conditions, such as metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD) cirrhosis, and obesity? (3) Does the siRNA used in previous studies knock down one or multiple transcripts? (4) Do different transcripts have the same or different functions for gene regulation? The presented data confirm the existence of several annotated HNF1A-AS1 transcripts in liver samples and cell lines, but also identify some new transcripts, which are not annotated in the Ensembl genome database. Expression patterns of the identified HNF1A-AS1 transcripts are highly correlated with the cell differentiation of matured hepatocyte-like cells from human embryonic stem cells (hESC), growth and differentiation of HepaRG cells, in response to rifampicin induction, and in various liver disease conditions. The expression levels of the HNF1A-AS1 transcripts are also highly correlated to the expression of cytochrome P450 enzymes, such as CYP3A4, during HepaRG growth, differentiation, and in response to rifampicin induction.

3.
Toxicol Sci ; 199(2): 316-331, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38526215

ABSTRACT

Bile acids (BAs) are signaling molecules synthesized in the liver initially by CYP7A1 and CYP27A1 in the classical and alternative pathways, respectively. BAs are essential for cholesterol clearance, intestinal absorption of lipids, and endogenous modulators of farnesoid x receptor (FXR). FXR is critical in maintaining BA homeostasis and gut-liver crosstalk. Complex reactions in vivo and the lack of suitable animal models impede our understanding of the functions of individual BAs. In this study, we characterized the in vivo effects of three-day feeding of cholic acid (CA), deoxycholic acid (DCA), or ursodeoxycholic acid (UDCA) at physiological/non-hepatotoxic concentrations in a novel low-BA mouse model (Cyp7a1-/-/Cyp27a1-/-, DKO). Liver injury, BA levels and composition and BA signaling by the FXR-fibroblast growth factor 15 (FGF15) axis were determined. Overall, higher basal inflammation and altered lipid metabolism in DKO mice might be associated with low BAs. CA, DCA, and UDCA feeding activated FXR signals with tissue specificity. Dietary CA and DCA similarly altered tissue BA profiles to be less hydrophobic, while UDCA promoted a more hydrophobic tissue BA pool with the profiles shifted toward non-12α-OH BAs and secondary BAs. However, UDCA did not offer any overt protective effects as expected. These findings allow us to determine the precise effects of individual BAs in vivo on BA-FXR signaling and overall BA homeostasis in liver physiology and pathologies.


Subject(s)
Bile Acids and Salts , Cholic Acid , Fibroblast Growth Factors , Liver , Mice, Knockout , Receptors, Cytoplasmic and Nuclear , Animals , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Bile Acids and Salts/metabolism , Liver/metabolism , Liver/drug effects , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Cholic Acid/metabolism , Male , Mice, Inbred C57BL , Deoxycholic Acid/toxicity , Cholestanetriol 26-Monooxygenase/genetics , Cholestanetriol 26-Monooxygenase/metabolism , Mice , Ursodeoxycholic Acid/pharmacology , Signal Transduction/drug effects , Cholesterol 7-alpha-Hydroxylase
4.
bioRxiv ; 2024 Jan 13.
Article in English | MEDLINE | ID: mdl-38260602

ABSTRACT

Background and Aims: Stearoyl-CoA desaturase-1 (SCD1) converts saturated fatty acids into monounsaturated fatty acids and plays an important regulatory role in lipid metabolism. Previous studies have demonstrated that mice deficient in SCD1 are protected from diet-induced obesity and hepatic steatosis due to altered lipid esterification and increased energy expenditure. Previous studies in our lab have shown that intestinal SCD1 modulates intestinal and plasma lipids and alters cholesterol metabolism. Here we investigated a novel role for intestinal SCD1 in the regulation of systemic energy balance. Methods: To interrogate the role of intestinal SCD1 in modulating whole body metabolism, intestine-specific Scd1 knockout (iKO) mice were maintained on standard chow diet or challenged with a high-fat diet (HFD). Studies included analyses of bile acid content and composition, metabolic phenotyping including body composition, indirect calorimetry, glucose tolerance analyses, and assessment of bile acid signaling pathways. Results: iKO mice displayed elevated plasma and hepatic bile acid content and decreased fecal bile acid excretion, associated with increased expression of the ileal bile acid uptake transporter, Asbt . These increases were associated with increased expression of TGR5 targets, including Dio2 in brown adipose tissue and elevated plasma glucagon-like peptide-1 levels. Upon HFD challenge, iKO mice had reduced metabolic efficiency apparent through decreased weight gain despite higher food intake. Concomitantly, energy expenditure was increased, and glucose tolerance was improved in HFD-fed iKO mice. Conclusion: Our results indicate that deletion of intestinal SCD1 has significant impacts on bile acid metabolism and whole-body energy balance, likely via activation of TGR5.

5.
Nat Commun ; 15(1): 627, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245529

ABSTRACT

Cancer cachexia is a systemic metabolic syndrome characterized by involuntary weight loss, and muscle and adipose tissue wasting. Mechanisms underlying cachexia remain poorly understood. Leukemia inhibitory factor (LIF), a multi-functional cytokine, has been suggested as a cachexia-inducing factor. In a transgenic mouse model with conditional LIF expression, systemic elevation of LIF induces cachexia. LIF overexpression decreases de novo lipogenesis and disrupts lipid homeostasis in the liver. Liver-specific LIF receptor knockout attenuates LIF-induced cachexia, suggesting that LIF-induced functional changes in the liver contribute to cachexia. Mechanistically, LIF overexpression activates STAT3 to downregulate PPARα, a master regulator of lipid metabolism, leading to the downregulation of a group of PPARα target genes involved in lipogenesis and decreased lipogenesis in the liver. Activating PPARα by fenofibrate, a PPARα agonist, restores lipid homeostasis in the liver and inhibits LIF-induced cachexia. These results provide valuable insights into cachexia, which may help develop strategies to treat cancer cachexia.


Subject(s)
Cachexia , Neoplasms , Animals , Mice , Cachexia/genetics , Cachexia/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Lipids , Lipogenesis/genetics , Liver/metabolism , Mice, Transgenic , Neoplasms/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism
6.
Mol Pharmacol ; 105(3): 179-193, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38238100

ABSTRACT

The circadian clock is an endogenous biochemical timing system that coordinates the physiology and behavior of organisms to earth's ∼24-hour circadian day/night cycle. The central circadian clock synchronized by environmental cues hierarchically entrains peripheral clocks throughout the body. The circadian system modulates a wide variety of metabolic signaling pathways to maintain whole-body metabolic homeostasis in mammals under changing environmental conditions. Endocrine fibroblast growth factors (FGFs), namely FGF15/19, FGF21, and FGF23, play an important role in regulating systemic metabolism of bile acids, lipids, glucose, proteins, and minerals. Recent evidence indicates that endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between peripheral clocks and energy homeostasis by regulating the expression of metabolic enzymes and hormones. Circadian disruption induced by environmental stressors or genetic ablation is associated with metabolic dysfunction and diurnal disturbances in FGF signaling pathways that contribute to the pathogenesis of metabolic diseases. Time-restricted feeding strengthens the circadian pattern of metabolic signals to improve metabolic health and prevent against metabolic diseases. Chronotherapy, the strategic timing of medication administration to maximize beneficial effects and minimize toxic effects, can provide novel insights into linking biologic rhythms to drug metabolism and toxicity within the therapeutical regimens of diseases. Here we review the circadian regulation of endocrine FGF signaling in whole-body metabolism and the potential effect of circadian dysfunction on the pathogenesis and development of metabolic diseases. We also discuss the potential of chrononutrition and chronotherapy for informing the development of timing interventions with endocrine FGFs to optimize whole-body metabolism in humans. SIGNIFICANCE STATEMENT: The circadian timing system governs physiological, metabolic, and behavioral functions in living organisms. The endocrine fibroblast growth factor (FGF) family (FGF15/19, FGF21, and FGF23) plays an important role in regulating energy and mineral metabolism. Endocrine FGFs function as nutrient sensors that mediate multifactorial interactions between circadian clocks and metabolic homeostasis. Chronic disruption of circadian rhythms increases the risk of metabolic diseases. Chronological interventions such as chrononutrition and chronotherapy provide insights into linking biological rhythms to disease prevention and treatment.


Subject(s)
Circadian Clocks , Metabolic Diseases , Humans , Animals , Circadian Rhythm/genetics , Circadian Clocks/genetics , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Metabolic Diseases/metabolism , Energy Metabolism , Mammals/metabolism
7.
J Pharmacol Exp Ther ; 388(2): 586-595, 2024 01 17.
Article in English | MEDLINE | ID: mdl-37188530

ABSTRACT

Nitrogen mustard (NM) is a cytotoxic vesicant known to cause pulmonary injury that can progress to fibrosis. NM toxicity is associated with an influx of inflammatory macrophages in the lung. Farnesoid X receptor (FXR) is a nuclear receptor involved in bile acid and lipid homeostasis that has anti-inflammatory activity. In these studies, we analyzed the effects of FXR activation on lung injury, oxidative stress, and fibrosis induced by NM. Male Wistar rats were exposed to phosphate-buffered saline (vehicle control) or NM (0.125 mg/kg) by intratracheal Penncentury-MicroSprayer aerosolization; this was followed by treatment with the FXR synthetic agonist, obeticholic acid (OCA, 15 mg/kg), or vehicle control (0.13-0.18 g peanut butter) 2 hours later and then once per day, 5 days per week thereafter for 28 days. NM caused histopathological changes in the lung, including epithelial thickening, alveolar circularization, and pulmonary edema. Picrosirius red staining and lung hydroxyproline content were increased, indicative of fibrosis; foamy lipid-laden macrophages were also identified in the lung. This was associated with aberrations in pulmonary function, including increases in resistance and hysteresis. Following NM exposure, lung expression of HO-1 and iNOS, and the ratio of nitrates/nitrites in bronchoalveolar lavage fluid (BAL), markers of oxidative stress increased, along with BAL levels of inflammatory proteins, fibrinogen, and sRAGE. Administration of OCA attenuated NM-induced histopathology, oxidative stress, inflammation, and altered lung function. These findings demonstrate that FXR plays a role in limiting NM-induced lung injury and chronic disease, suggesting that activating FXR may represent an effective approach to limiting NM-induced toxicity. SIGNIFICANCE STATEMENT: In this study, the role of farnesoid-X-receptor (FXR) in mustard vesicant-induced pulmonary toxicity was analyzed using nitrogen mustard (NM) as a model. This study's findings that administration of obeticholic acid, an FXR agonist, to rats reduces NM-induced pulmonary injury, oxidative stress, and fibrosis provide novel mechanistic insights into vesicant toxicity, which may be useful in the development of efficacious therapeutics.


Subject(s)
Chenodeoxycholic Acid/analogs & derivatives , Lung Injury , Mechlorethamine , Rats , Male , Animals , Mechlorethamine/toxicity , Irritants/adverse effects , Rats, Wistar , Lung , Fibrosis , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/pathology , Lung Injury/metabolism , Oxidative Stress , Lipids
8.
Hepatobiliary Pancreat Dis Int ; 22(5): 466-473, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37620226

ABSTRACT

The synthesis of bile acids (BAs) is carried out by complex pathways characterized by sequential chemical reactions in the liver through various cytochromes P450 (CYP) and other enzymes. Maintaining the integrity of these pathways is crucial for normal physiological function in mammals, encompassing hepatic and neurological processes. Studying on the deficiencies in BA synthesis genes offers valuable insights into the significance of BAs in modulating farnesoid X receptor (FXR) signaling and metabolic homeostasis. By creating mouse knockout (KO) models, researchers can manipulate deficiencies in genes involved in BA synthesis, which can be used to study human diseases with BA dysregulation. These KO mouse models allow for a more profound understanding of the functions and regulations of genes responsible for BA synthesis. Furthermore, KO mouse models shed light on the distinct characteristics of individual BA and their roles in nuclear receptor signaling. Notably, alterations of BA synthesis genes in mouse models have distinct differences when compared to human diseases caused by the same BA synthesis gene deficiencies. This review summarizes several mouse KO models used to study BA synthesis and related human diseases, including mice deficient in Cyp7a1, Cyp27a1, Cyp7a1/Cyp27a1, Cyp8b1, Cyp7b1, Cyp2c70, Cyp2a12, and Cyp2c70/Cyp2a12, as well as germ-free mice.


Subject(s)
Bile Acids and Salts , Liver , Mice , Humans , Animals , Liver/metabolism , Bile Acids and Salts/metabolism , Disease Models, Animal , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Mice, Inbred C57BL , Mammals
9.
Semin Liver Dis ; 43(3): 267-278, 2023 08.
Article in English | MEDLINE | ID: mdl-37442156

ABSTRACT

Chronic liver diseases encompass a wide spectrum of hepatic maladies that often result in cholestasis or altered bile acid secretion and regulation. Incidence and cost of care for many chronic liver diseases are rising in the United States with few Food and Drug Administration-approved drugs available for patient treatment. Farnesoid X receptor (FXR) is the master regulator of bile acid homeostasis with an important role in lipid and glucose metabolism and inflammation. FXR has served as an attractive target for management of cholestasis and fibrosis; however, global FXR agonism results in adverse effects in liver disease patients, severely affecting quality of life. In this review, we highlight seminal studies and recent updates on the FXR proteome and identify gaps in knowledge that are essential for tissue-specific FXR modulation. In conclusion, one of the greatest unmet needs in the field is understanding the underlying mechanism of intestinal versus hepatic FXR function.


Subject(s)
Cholestasis , Liver Diseases , Humans , Friends , Quality of Life , Liver/metabolism , Cholestasis/drug therapy , Liver Diseases/drug therapy , Liver Diseases/metabolism , Bile Acids and Salts/metabolism
10.
Mol Cancer ; 22(1): 110, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443114

ABSTRACT

BACKGROUND: Drugs targeting the spindle assembly checkpoint (SAC), such as inhibitors of Aurora kinase B (AURKB) and dual specific protein kinase TTK, are in different stages of clinical development. However, cell response to SAC abrogation is poorly understood and there are no markers for patient selection. METHODS: A panel of 53 tumor cell lines of different origins was used. The effects of drugs were analyzed by MTT and flow cytometry. Copy number status was determined by FISH and Q-PCR; mRNA expression by nCounter and RT-Q-PCR and protein expression by Western blotting. CRISPR-Cas9 technology was used for gene knock-out (KO) and a doxycycline-inducible pTRIPZ vector for ectopic expression. Finally, in vivo experiments were performed by implanting cultured cells or fragments of tumors into immunodeficient mice. RESULTS: Tumor cells and patient-derived xenografts (PDXs) sensitive to AURKB and TTK inhibitors consistently showed high expression levels of BH3-interacting domain death agonist (BID), while cell lines and PDXs with low BID were uniformly resistant. Gene silencing rendered BID-overexpressing cells insensitive to SAC abrogation while ectopic BID expression in BID-low cells significantly increased sensitivity. SAC abrogation induced activation of CASP-2, leading to cleavage of CASP-3 and extensive cell death only in presence of high levels of BID. Finally, a prevalence study revealed high BID mRNA in 6% of human solid tumors. CONCLUSIONS: The fate of tumor cells after SAC abrogation is driven by an AURKB/ CASP-2 signaling mechanism, regulated by BID levels. Our results pave the way to clinically explore SAC-targeting drugs in tumors with high BID expression.


Subject(s)
Neoplasms , Protein Serine-Threonine Kinases , Humans , Animals , Mice , Protein Serine-Threonine Kinases/genetics , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , M Phase Cell Cycle Checkpoints , Cell Line, Tumor , RNA, Messenger , Neoplasms/drug therapy , Neoplasms/genetics , Protein-Tyrosine Kinases/metabolism , Cell Cycle Proteins/genetics
11.
Hepatol Commun ; 7(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37058105

ABSTRACT

NASH is within the spectrum of NAFLD, a liver condition encompassing liver steatosis, inflammation, hepatocyte injury, and fibrosis. The prevalence of NASH-induced cirrhosis is rapidly rising and has become the leading indicator for liver transplantation in the US. There is no Food and Drug Administration (FDA)-approved pharmacological intervention for NASH. The farnesoid X receptor (FXR) is essential in regulating bile acid homeostasis, and dysregulation of bile acids has been implicated in the pathogenesis of NASH. As a result, modulators of FXR that show desirable effects in mitigating key characteristics of NASH have been developed as promising therapeutic approaches. However, global FXR activation causes adverse effects such as cholesterol homeostasis imbalance and pruritus. The development of targeted FXR modulation is necessary for ideal NASH therapeutics, but information regarding tissue-specific and cell-specific FXR functionality is limited. In this review, we highlight FXR activation in the regulation of bile acid homeostasis and NASH development, examine the current literature on tissue-specific regulation of nuclear receptors, and speculate on how FXR regulation will be beneficial in the treatment of NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/pathology , Liver Cirrhosis , Bile Acids and Salts , Receptors, Cytoplasmic and Nuclear , Inflammation
12.
Hepatology ; 77(4): 1164-1180, 2023 04 01.
Article in English | MEDLINE | ID: mdl-35689610

ABSTRACT

BACKGROUND AND AIMS: Intestinal farnesoid X receptor (FXR) plays a critical role in alcohol-associated liver disease (ALD). We aimed to investigate whether alcohol-induced dysbiosis increased intestinal microRNA194 (miR194) that suppressed Fxr transcription and whether Lactobacillus rhamnosus GG-derived exosome-like nanoparticles (LDNPs) protected against ALD through regulation of intestinal miR194-FXR signaling in mice. APPROACH AND RESULTS: Binge-on-chronic alcohol exposure mouse model was utilized. In addition to the decreased ligand-mediated FXR activation, alcohol feeding repressed intestinal Fxr transcription and increased miR194 expression. This transcriptional suppression of Fxr by miR194 was confirmed in intestinal epithelial Caco-2 cells and mouse enteriods. The alcohol feeding-reduced intestinal FXR activation was further demonstrated by the reduced FXR reporter activity in fecal samples and by the decreased fibroblast growth factor 15 (Fgf15) messenger RNA (mRNA) in intestine and protein levels in the serum, which caused an increased hepatic bile acid synthesis and lipogeneses. We further demonstrated that alcohol feeding increased-miR194 expression was mediated by taurine-upregulated gene 1 (Tug1) through gut microbiota regulation of taurine metabolism. Importantly, 3-day oral administration of LDNPs increased bile salt hydrolase (BSH)-harboring bacteria that decreased conjugated bile acids and increased gut taurine concentration, which upregulated Tug1, leading to a suppression of intestinal miR194 expression and recovery of FXR activation. Activated FXR upregulated FGF15 signaling and subsequently reduced hepatic bile acid synthesis and lipogenesis and attenuated ALD. These protective effects of LDNPs were eliminated in intestinal FxrΔIEC and Fgf15-/- mice. We further showed that miR194 was upregulated, whereas BSH activity and taurine levels were decreased in fecal samples of patients with ALD. CONCLUSIONS: Our results demonstrated that gut microbiota-mediated miR194 regulation contributes to ALD pathogenesis and to the protective effects of LDNPs through modulating intestinal FXR signaling.


Subject(s)
Liver Diseases, Alcoholic , MicroRNAs , Animals , Humans , Mice , Bile Acids and Salts/metabolism , Caco-2 Cells , Ethanol/pharmacology , Liver/pathology , Liver Diseases, Alcoholic/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Taurine/pharmacology , Nanoparticles
14.
Toxicol Sci ; 191(2): 387-399, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36511616

ABSTRACT

The effects of exposure to Myclobutanil, a triazole fungicide, on the development and progression of nonalcoholic fatty liver disease (NAFLD) are unclear, but activation of nuclear receptors (NRs) is a known mechanism of azole-induced liver toxicity. Farnesoid X receptor (FXR) is a NR and is highly expressed in the liver and intestine. Activation of FXR tightly regulates bile acid (BA), lipid and glucose homeostasis, and inflammation partly through the induction of fibroblast growth factor 15 (FGF15; human ortholog FGF19). FXR activation is downregulated during NAFLD and agonists are currently being explored as potential therapeutic strategy. In this study, we aimed to clarify the effects of Myclobutanil exposure on FXR activation and NAFLD development. Reporter assay showed Myclobutanil treatment, following FXR activation with potent FXR agonist (GW4064), resulted in a dose-dependent decrease of FXR activity. Furthermore, a 10-day study in male mice demonstrated that cotreatment with Myclobutanil led to an 80% reduction of GW4064-induced ileal expression of Fgf15. In a diet-induced NAFLD study, low-fat diet (LFD) fed mice administered myclobutanil displayed decreased FXR activity in the liver and ileum, while high-fat-high-sugar-diet (HFHSD) fed mice showed an increase in hepatic FXR activity and an induction of target genes regulated by constitutive androstane receptor and/or pregnane X receptor. Our work demonstrates Myclobutanil inhibits FXR activity and modulates FXR activity differentially in mice fed LFD or HFHSD. Our studies suggest the importance of understanding how Myclobutanil could contribute to BA dysregulation in disease states such as NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Triazoles , Animals , Humans , Male , Mice , Bile Acids and Salts/metabolism , Intestines/metabolism , Liver/metabolism , Mice, Inbred C57BL , Nitriles/pharmacology , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/metabolism , RNA-Binding Proteins/metabolism , Triazoles/toxicity , Triazoles/metabolism
15.
Toxicol Appl Pharmacol ; 454: 116208, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35998709

ABSTRACT

Nitrogen mustard (NM) is a cytotoxic vesicant known to cause acute lung injury which progresses to fibrosis; this is associated with a sequential accumulation of pro- and anti-inflammatory macrophages in the lung which have been implicated in NM toxicity. Farnesoid X receptor (FXR) is a nuclear receptor involved in regulating lipid homeostasis and inflammation. In these studies, we analyzed the role of FXR in inflammatory macrophage activation, lung injury and oxidative stress following NM exposure. Wild-type (WT) and FXR-/- mice were treated intratracheally with PBS (control) or NM (0.08 mg/kg). Bronchoalveolar lavage fluid (BAL) and lung tissue were collected 3, 14 and 28 d later. NM caused progressive histopathologic alterations in the lung including inflammatory cell infiltration and alveolar wall thickening and increases in protein and cells in BAL; oxidative stress was also noted, as reflected by upregulation of heme oxygenase-1. These changes were more prominent in male FXR-/- mice. Flow cytometric analysis revealed that loss of FXR resulted in increases in proinflammatory macrophages at 3 d post NM; this correlated with upregulation of COX-2 and ARL11, markers of macrophage activation. Markers of anti-inflammatory macrophage activation, CD163 and STAT6, were also upregulated after NM; this response was exacerbated in FXR-/- mice at 14 d post-NM. These findings demonstrate that FXR plays a role in limiting macrophage inflammatory responses important in lung injury and oxidative stress. Maintaining or enhancing FXR function may represent a useful strategy in the development of countermeasures to treat mustard lung toxicity.


Subject(s)
Acute Lung Injury , Mechlorethamine , Acute Lung Injury/pathology , Animals , Cyclooxygenase 2/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Irritants/toxicity , Lipids , Lung , Macrophage Activation , Male , Mechlorethamine/toxicity , Mice
16.
EMBO J ; 41(17): e109997, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35686465

ABSTRACT

Lysosome-mediated macroautophagy, including lipophagy, is activated under nutrient deprivation but is repressed after feeding. We show that, unexpectedly, feeding activates intestinal autophagy/lipophagy in a manner dependent on both the orphan nuclear receptor, small heterodimer partner (SHP/NR0B2), and the gut hormone, fibroblast growth factor-15/19 (FGF15/19). Furthermore, postprandial intestinal triglycerides (TGs) and apolipoprotein-B48 (ApoB48), the TG-rich chylomicron marker, were elevated in SHP-knockout and FGF15-knockout mice. Genomic analyses of the mouse intestine indicated that SHP partners with the key lysosomal activator, transcription factor-EB (TFEB) to upregulate the transcription of autophagy/lipolysis network genes after feeding. FGF19 treatment activated lipophagy, reducing TG and ApoB48 levels in HT29 intestinal cells, which was dependent on TFEB. Mechanistically, feeding-induced FGF15/19 signaling increased the nuclear localization of TFEB and SHP via PKC beta/zeta-mediated phosphorylation, leading to increased transcription of the TFEB/SHP target lipophagy genes, Ulk1 and Atgl. Collectively, these results demonstrate that paradoxically after feeding, FGF15/19-activated SHP and TFEB activate gut lipophagy, limiting postprandial TGs. As excess postprandial lipids cause dyslipidemia and obesity, the FGF15/19-SHP-TFEB axis that reduces intestinal TGs via lipophagic activation provides promising therapeutic targets for obesity-associated metabolic disease.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Eating , Fibroblast Growth Factors , Gastrointestinal Tract , Receptors, Cytoplasmic and Nuclear , Animals , Apolipoprotein B-48/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fibroblast Growth Factors/metabolism , Gastrointestinal Tract/metabolism , Lysosomes/metabolism , Mice , Mice, Knockout , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
17.
Am J Transl Res ; 14(2): 1092-1099, 2022.
Article in English | MEDLINE | ID: mdl-35273712

ABSTRACT

OBJECTIVE: Mortality-trends from alcoholic liver disease (ALD) have recently increased and they differ by various factors in the U.S. However, these trends have only been analyzed using univariate models and in reality they may be influenced by various factors. We thus examined trends in age-standardized mortality from ALD among U.S. adults for 1999-2017, using multivariable piecewise log-linear models. METHODS: We collected mortality-data from the Centers for Disease Control and Prevention Wide-ranging Online Data for Epidemiologic Research database, using the Underlying Cause of Death. RESULTS: We identified 296,194 deaths from ALD and 346,386 deaths indirectly attributable to ALD during the period from 1999-2017. The multivariable-adjusted, age-standardized ALD mortality was stable during 1999-2006 (annual percentage change [APC]=-2.24, P=0.24), and increased during 2006-2017 (APC=3.18, P<0.006). Their trends did not differ by sex, race, age or urbanization. Subgroup analyses revealed upward multivariable-adjusted, age-standardized mortality-trends in alcoholic fatty liver (APC=4.64, P<0.001), alcoholic hepatitis (APC=4.38, P<0.001), and alcoholic cirrhosis (APC=5.33, P<0.001), but downward mortality-trends in alcoholic hepatic failure (APC=-1.63, P=0.006) and unspecified ALD (APC=-0.86, P=0.013). Strikingly, non-alcoholic cirrhosis also had an upward multivariable-adjusted, age-standardized mortality-trend (APC=0.69, P=0.046). By contrast, recent mortality-trends were stable for all cause of deaths (APC=-0.39, P=0.379) and downward for malignant neoplasms excluding liver cancer (APC=-2.82, P<0.001), infections (APC=-2.60, P<0.001), cardiovascular disease (APC=-0.69, P=0.044) and respiratory disease (APC=-0.56, P=0.002). The adjusted mortality with ALD as a contributing cause of death also had an upward trend during 2000-2017 (APC=5.47, P<0.001). Strikingly, common comorbidities of ALD, including hepatocellular carcinoma, cerebrovascular and ischemic heart cardiovascular diseases and sepsis, had upward trends during the past 14 to 16 years. CONCLUSIONS: ALD had an upward multivariable-adjusted, age-standardized mortality-trend among U.S. adults, without significant differences by sex, race, age or urbanization. Three ALD subtypes (alcoholic fatty liver, alcoholic hepatitis and alcoholic cirrhosis) and non-alcoholic cirrhosis had upward morality-trends, while other ALD subtypes and other causes of death did not.

18.
J Clin Invest ; 132(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35349482

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), the most common liver disease, has become a silent worldwide pandemic. The incidence of NAFLD correlates with the rise in obesity, type 2 diabetes, and metabolic syndrome. A hallmark featureof NAFLD is excessive hepatic fat accumulation or steatosis, due to dysregulated hepatic fat metabolism, which can progress to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Currently, there are no approved pharmacotherapies to treat this disease. Here, we have found that activation of the kisspeptin 1 receptor (KISS1R) signaling pathway has therapeutic effects in NAFLD. Using high-fat diet-fed mice, we demonstrated that a deletion of hepatic Kiss1r exacerbated hepatic steatosis. In contrast, enhanced stimulation of KISS1R protected against steatosis in wild-type C57BL/6J mice and decreased fibrosis using a diet-induced mouse model of NASH. Mechanistically, we found that hepatic KISS1R signaling activates the master energy regulator, AMPK, to thereby decrease lipogenesis and progression to NASH. In patients with NAFLD and in high-fat diet-fed mice, hepatic KISS1/KISS1R expression and plasma kisspeptin levels were elevated, suggesting a compensatory mechanism to reduce triglyceride synthesis. These findings establish KISS1R as a therapeutic target to treat NASH.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Kisspeptins/genetics , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Kisspeptin-1/genetics , Receptors, Kisspeptin-1/metabolism
19.
Methods Mol Biol ; 2455: 305-318, 2022.
Article in English | MEDLINE | ID: mdl-35213003

ABSTRACT

Bile acids (BAs) serve as important signaling molecules and are endogenous ligands of nuclear and cell membrane receptors to regulate physiological and pathological processes. BA synthesis and metabolism have been impaired in NASH patients because of liver injury, inflammation or obstruction of bile ducts. On the other hand, the changes in BA composition might alter the activation status of various cell signaling pathways and contribute to NASH pathogenesis. Due to the rapidly increasing interests in the roles of individual BA in disease development, this chapter will focus on the method for analyzing individual BA profile in mouse biofluids and tissues by high-performance liquid chromatography coupled with ion trap mass spectrometry (HPLC-MS).


Subject(s)
Bile Acids and Salts , Liver , Animals , Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Chromatography, High Pressure Liquid , Humans , Liver/metabolism , Mass Spectrometry , Mice
20.
J Clin Exp Hepatol ; 12(1): 200-203, 2022.
Article in English | MEDLINE | ID: mdl-35068799

ABSTRACT

Bile acid metabolism is altered in neonates on parenteral nutrition (PN), predisposing them to parenteral nutrition-associated liver disease. Cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in the bile acid synthesis pathway, is repressed by fibroblast growth factor 19 (FGF19) and phytosterols (PS). We describe a case of a preterm infant who developed necrotizing enterocolitis (NEC) and received exclusive PN for over 2 months. Our objective was to serially assess CYP7A1 activity and plasma FGF19 and PS concentrations in this infant case compared to five healthy preterm infants. We found that CYP7A1 activity increased during the first 2 weeks of life in control infants but was undetectable in the infant case. FGF19 concentrations were high at birth in all infants and subsequently declined and did not differ between the case and control infants. As expected, PS concentrations were elevated in the infant case and continued to increase despite lipid minimization. In conclusion, CYP7A1 activity was gradually upregulated in healthy preterm infants but remained suppressed in the infant requiring prolonged PN. Preterm infants also had elevated FGF19 concentrations at birth, which decreased with advancing postnatal age.

SELECTION OF CITATIONS
SEARCH DETAIL
...