1
|
Cao H, Wang Z, Xu L, Han B, Sun D. Genetic Associations of ACOX2 Gene with Milk Yield and Composition Traits in Chinese Holstein Cows. Animals (Basel) 2025; 15:953. [PMID: 40218347 PMCID: PMC11987739 DOI: 10.3390/ani15070953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
In our previous studies on the liver proteome of Holstein cows, the acyl-CoA oxidase 2 (ACOX2) gene was identified as a promising candidate for milk traits, being involved in the processes of fatty acid metabolism and bile acid formation. Herein, we evaluated its genetic effects on milk production traits in 922 Chinese Holstein cows. By sequencing the entire coding region and 2000 bp of the 5' and 3' flanking sequences of the ACOX2 gene, we identified a total of five SNPs, including one SNP in the 5' UTR, one in intron 5, and three in the 3' flanking region. Using an animal model, we found that the SNPs rs109066086, rs109665171, and rs454339362 were significantly associated with at least one of the milk production traits, including 305-day milk yield, milk fat yield, milk protein yield, milk fat percentage, and milk protein percentage in the first lactation (p ≤ 4.03 × 10-2). And in the second lactation, all five SNPs were significantly associated with at least three of the milk production traits (p ≤ 1.17 × 10-2). We also found that in the second lactation, the SNP rs209677248 had a high phenotypic variance rate for milk protein percentage, with a value of 4.90%. With Haploview 4.2, it was observed that the four SNPs formed two haplotype blocks, which were significantly associated with the 305-day milk, fat, and protein yields (p ≤ 1.03 × 10-2; p ≤ 8.60 × 10-3; p ≤ 3.20 × 10-3). In addition, it was predicted that the T allele in the SNP rs109066086 created TFBSs for transcription factors NC2R2 and TFAP4, thereby potentially affecting ACOX2 expression. Overall, our results provide the first confirmation of the genetic effects of the ACOX2 gene on milk yield and composition traits in dairy cattle and revealed the referable molecular markers for genomic selection.
Collapse
Affiliation(s)
| | | | | | | | - Dongxiao Sun
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal Genetics, National Engineering Laboratory of Animal Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Beijing 100193, China; (H.C.); (Z.W.); (L.X.); (B.H.)
| |
Collapse
|
2
|
Vaz FM, Ferdinandusse S, Salomons GS, Wanders RJA. Disorders of fatty acid homeostasis. J Inherit Metab Dis 2025; 48:e12734. [PMID: 38693715 PMCID: PMC11730842 DOI: 10.1002/jimd.12734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 05/03/2024]
Abstract
Humans derive fatty acids (FA) from exogenous dietary sources and/or endogenous synthesis from acetyl-CoA, although some FA are solely derived from exogenous sources ("essential FA"). Once inside cells, FA may undergo a wide variety of different modifications, which include their activation to their corresponding CoA ester, the introduction of double bonds, the 2- and ω-hydroxylation and chain elongation, thereby generating a cellular FA pool which can be used for the synthesis of more complex lipids. The biological properties of complex lipids are very much determined by their molecular composition in terms of the FA incorporated into these lipid species. This immediately explains the existence of a range of genetic diseases in man, often with severe clinical consequences caused by variants in one of the many genes coding for enzymes responsible for these FA modifications. It is the purpose of this review to describe the current state of knowledge about FA homeostasis and the genetic diseases involved. This includes the disorders of FA activation, desaturation, 2- and ω-hydroxylation, and chain elongation, but also the disorders of FA breakdown, including disorders of peroxisomal and mitochondrial α- and β-oxidation.
Collapse
Affiliation(s)
- Frédéric M. Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
| | - Gajja S. Salomons
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| | - Ronald J. A. Wanders
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic DiseasesEmma Children's Hospital, Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
- Inborn Errors of Metabolism, Amsterdam Gastroenterology Endocrinology MetabolismAmsterdamThe Netherlands
| |
Collapse
|
3
|
Feng Y, Liu CH, Yang J, Zhang H, Li L, Yang Q, Gan W, Yang Z, Gong P, Fu C, Qian G, Li D. Integrative analysis of non12-hydroxylated bile acid revealed the suppressed molecular map of alternative pathway in nonalcoholic steatohepatitis mice. FASEB J 2024; 38:e70167. [PMID: 39556333 DOI: 10.1096/fj.202401630r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
Bile acids (BAs) are significantly altered in the liver and serum of patients with nonalcoholic steatohepatitis (NASH). However, the underlying mechanisms of these changes, particularly BA alternative pathways (BAP) responsible for non12-OH BAs, remain unclear. RNA-seq data were initially analyzed to reveal the changes of gene expression in NASH patients. Targeted metabolomics were conducted on plasma from NASH mice induced by high-fat or western diet with CCl4 for 10-24 weeks. Liver tissues were examined using proteomics, RT-qPCR, and western blotting. An integrated approach was then employed to analyze protein interactions and network correlations. Analysis of RNA-seq data revealed the inhibition of CYP7B1 in NASH patients, indicating the dysregulation of BAP. In NASH mouse models, dysregulation of BA circulation was observed by increased plasma total BA (TBA) levels and decreased liver TBA, with liver swelling and histopathological changes. Targeted metabolomics revealed suppressed levels of non12-OH BAs, which inversely correlated with increased liver injury markers. The reduced mRNA and protein expression of Fxr and upregulation of Lxr signaling in livers suggested the suppressed BAP was modulated by Fxr-Lxr signaling. Moreover, BAP interactions predominantly implicated multiple metabolism disruptions, involving 7 hub proteins (Hk1, Acadsb, Pklr, Insr, Ldlr, Cyp27a1, and Cyp7b1), offering promising therapeutic targets for NASH. We presented the metabolic and proteomic map of BAP and its regulatory network in NASH progression. Therapeutic targeting of BAP or its co-regulatory proteins holds promise for NASH treatment and metabolic syndrome management.
Collapse
Affiliation(s)
- Yanruyu Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- Ninth People's Hospital of Zhengzhou, Zhengzhou, China
| | - Chang-Hai Liu
- Center of Infectious Diseases, Division of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jingtao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qian Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Wei Gan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zi Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Chunmei Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Guangsheng Qian
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Dapeng Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Sümer Coşar Ö, Öztürk H, Sarı S, Vilarinho S, Kayhan G, Eğritaş Gürkan Ö, Dalgıç B. Variable Clinical Spectrum of Inborn Errors of Bile Acid Synthesis: A Report of 10 Cases. EXP CLIN TRANSPLANT 2024; 22:100-105. [PMID: 39498929 DOI: 10.6002/ect.pedsymp2024.o29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
OBJECTIVES Inborn errors of bile acid synthesis are rare genetic disorders that usually present as neonatal cholestasis and liver disease in older children and adults. The symptomatology of inborn errors of bile acid synthesis can markedly vary among individuals, ranging from mild to severe conditions. Diagnosis is based on genetic tests and/or urine liquid secondary ionization mass spectrometry. Here, we have described characteristics of patients who were diagnosed with inborn errors of bile acid synthesis in our department. MATERIALS AND METHODS We retrospectively evaluated data from patients diagnosed with inborn errors of bile acid synthesis by urine bile acid analysis and/or genetic tests between 2013 and 2023. RESULTS Ten patients (8 boys, 2 girls) born to consanguineous parents were diagnosed with inborn errors of bile acid synthesis during the study period. Six patients were diagnosed with 3β-hydroxy-Δ5-C27-steroid dehydrogenase deficiency, 2 patients with peroxisomal acyl-CoA oxidase 2 deficiency, and 2 patients with peroxisome biogenesis disorder. In patients with 3β-hydroxy-Δ5-C27-steroid dehydrogenase deficiency, 3 patients were monitored with cholic acid treatment, 2 underwent liver transplant due to liver failure, and 1 patient died from liver failure. Ursodeoxycholic acid treatment was given to patients with acyl-CoA oxidase 2 deficiency. Cholic acid was given to patients with peroxisome biogenesis disorder. CONCLUSIONS Inborn errors of bile acid synthesis can cause a variety of liver diseases, from asymptomatic liver enzyme elevation to cirrhosis. Clinical findings may include neurological symptoms and fat and fat-soluble vitamin malabsorption complications. Deficiency of 3β-hydroxy-Δ5-C27-steroid dehydrogenase is the most common bile acid synthetic defect presenting in cholestasis in infancy and childhood. Cholic acid is effective for most patients with inborn errors of bile acid synthesis. If patients do not receive an early diagnosis, progressive liver disease or other serious complications may develop.
Collapse
Affiliation(s)
- Özlem Sümer Coşar
- From Gazi University School of Medicine, Department of Pediatric Gastroenterology Hepatology and Nutrition, Ankara, Turkey
| | | | | | | | | | | | | |
Collapse
|
5
|
Guerra-Ruiz A. Kick-start for metabolomics in liver disease. World J Hepatol 2024; 16:1206-1210. [PMID: 39351520 PMCID: PMC11438589 DOI: 10.4254/wjh.v16.i9.1206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 09/23/2024] Open
Abstract
It is not complicated for the clinician to diagnose a patient with advanced fibrosis or liver cirrhosis when he has already presented some decompensation of his liver disease. However, it is in the earliest stages when the patient's prognosis can be modified the most. Since liver disease is generally asymptomatic, not invasive markers are of great relevance. In the era of omics, it is time for metabolomics to accompany genomics and proteomics, which are more established in the diagnostics and prognostics clinical toolbox. Metabolomics, understood as the comprehensive evaluation of the metabolites present in the organism in a specific physiological situation, has undoubted advantages in the study and identification of serum markers relevant to a specific pathology. Last year, I read with interest two articles published in this journal: "Baseline metabolites could predict responders with hepatitis B virus-related liver fibrosis for entecavir or combined with FuzhengHuayu tablet" by Dai et al and "Metabolomics in chronic hepatitis C: Decoding fibrosis grading and underlying pathways" by Ferrasi et al. Both papers illuminate the power of metabolomics to provide us with new tools in the management of liver disease. In this editorial, I comment on these studies and others, and note how they can contribute to our understanding of liver disease in more than one way.
Collapse
Affiliation(s)
- Armando Guerra-Ruiz
- Laboratory Medicine, University Hospital Marques de Valdecilla, Santander 39008, Cantabria, Spain
- Commission on Biochemical Assessment of Liver Disease, Spanish Society of Clinical Chemistry (SEQC-ML), Barcelona 08025, Catalonia, Spain
- Digestive Diseases, Valdecilla Research Institute (IDIVAL), Santander 39011, Cantabria, Spain.
| |
Collapse
|
6
|
Lülecioğlu AA, Yazıcı YY, Baran A, Warasnhe K, Beyaz Ş, Aytekin C, Özçay F, Aydemir Y, Barış Z, Belkaya S. Whole-exome sequencing for genetic diagnosis of idiopathic liver injury in children. J Cell Mol Med 2024; 28:e18485. [PMID: 38864694 PMCID: PMC11167704 DOI: 10.1111/jcmm.18485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Genome-wide approaches, such as whole-exome sequencing (WES), are widely used to decipher the genetic mechanisms underlying inter-individual variability in disease susceptibility. We aimed to dissect inborn monogenic determinants of idiopathic liver injury in otherwise healthy children. We thus performed WES for 20 patients presented with paediatric-onset recurrent elevated transaminases (rELT) or acute liver failure (ALF) of unknown aetiology. A stringent variant screening was undertaken on a manually-curated panel of 380 genes predisposing to inherited human diseases with hepatobiliary involvement in the OMIM database. We identified rare nonsynonymous variants in nine genes in six patients (five rELT and one ALF). We next performed a case-level evaluation to assess the causal concordance between the gene mutated and clinical symptoms of the affected patient. A genetic diagnosis was confirmed in four rELT patients (40%), among whom two carried novel mutations in ACOX2 or PYGL, and two had previously-reported morbid variants in ABCB4 or PHKA2. We also detected rare variants with uncertain clinical significance in CDAN1, JAG1, PCK2, SLC27A5 or VPS33B in rELT or ALF patients. In conclusion, implementation of WES improves diagnostic yield and enables precision management in paediatric cases of liver injury with unknown aetiology, in particular recurrent hypertransaminasemia.
Collapse
Affiliation(s)
- Aysima Atılgan Lülecioğlu
- Department of Molecular Biology and Genetics, Faculty of Scienceİhsan Doğramacı Bilkent UniversityAnkaraTurkey
| | - Yılmaz Yücehan Yazıcı
- Department of Molecular Biology and Genetics, Faculty of Scienceİhsan Doğramacı Bilkent UniversityAnkaraTurkey
| | - Alperen Baran
- Department of Molecular Biology and Genetics, Faculty of Scienceİhsan Doğramacı Bilkent UniversityAnkaraTurkey
| | - Khaled Warasnhe
- Department of PediatricsBaşkent University Faculty of MedicineAnkaraTurkey
| | - Şengül Beyaz
- Department of Immunology and Allergy DiseasesAnkara Bilkent City HospitalAnkaraTurkey
| | - Caner Aytekin
- Department of Pediatric ImmunologyDr. Sami Ulus Maternity and Children's Health and Diseases Training and Research HospitalAnkaraTurkey
| | - Figen Özçay
- Department of Pediatric Gastroenterology and HepatologyBaşkent University Faculty of MedicineAnkaraTurkey
| | - Yusuf Aydemir
- Department of Pediatric Gastroenterology, Faculty of MedicineEskişehir Osmangazi UniversityEskişehirTurkey
| | - Zeren Barış
- Department of Pediatric Gastroenterology, Faculty of MedicineEskişehir Osmangazi UniversityEskişehirTurkey
| | - Serkan Belkaya
- Department of Molecular Biology and Genetics, Faculty of Scienceİhsan Doğramacı Bilkent UniversityAnkaraTurkey
| |
Collapse
|
7
|
Han J, Wu P, Wen Y, Liu C, Liu X, Tao H, Zhang F, Zhang X, Ye Q, Shen T, Chen X, Yu H. The zhuyu pill relieves rat cholestasis by regulating the mRNA expression of lipid and bile metabolism associated genes. Front Pharmacol 2023; 14:1280864. [PMID: 37881184 PMCID: PMC10597705 DOI: 10.3389/fphar.2023.1280864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023] Open
Abstract
Background: The Zhuyu pill (ZYP), composed of Coptis chinensis Franch. and Tetradium ruticarpum (A. Jussieu) T. G. Hartley, is an effective traditional Chinese medicine with potential anti-cholestatic effects. However, the underlying mechanisms of ZYP remain unknown. Objective: To investigate the mechanism underlying the interventional effect of ZYP on mRNA-seq analysis in cholestasis rat models. Materials and methods: This study tested the effects of a low-dose (0.6 g/kg) and high-dose (1.2 g/kg) of ZYP on a cholestasis rat model induced by α-naphthyl-isothiocyanate (ANIT, 50 mg/kg). Serum biochemistry and histopathology results were used to evaluate the therapeutic effect of ZYP, and mRNA-Seq analysis was performed and verified using real-time fluorescence quantitative PCR (qRT-PCR). GO, KEGG, and GSEA analyses were integrated to identify the mechanism by which ZYP impacted cholestatic rats. Results: ZYP was shown to significantly improve abnormal changes in the biochemical blood indexes and liver histopathology of cholestasis rats and regulate pathways related to bile and lipid metabolism, including fatty acid metabolism, retinol metabolism, and steroid hormone biosynthesis, to alleviate inflammation, cholestasis, and lipid metabolism disorders. Relative expression of the essential genes Cyp2a1, Ephx2, Acox2, Cyp1a2, Cyp2c11, and Sult2a1 was verified by qRT-PCR and showed the same trend as mRNA-seq analysis. Conclusion: ZYP has a significant anti-cholestatic effect by regulating bile metabolism and lipid metabolism related pathways. These findings indicate that ZYP is a novel and promising prospect for treating cholestasis.
Collapse
Affiliation(s)
- Jun Han
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peijie Wu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pediatrics, Guang’an Traditional Chinese Medicine Hospital, Guang’an, China
| | - Chao Liu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinglong Liu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Tao
- Department of Dermatology, Cangxi Traditional Chinese Medicine Hospital, Guangyuan, China
| | - Fenghua Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaodan Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiaobo Ye
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofeng Chen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Ranea-Robles P, Houten SM. The biochemistry and physiology of long-chain dicarboxylic acid metabolism. Biochem J 2023; 480:607-627. [PMID: 37140888 PMCID: PMC10214252 DOI: 10.1042/bcj20230041] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/05/2023]
Abstract
Mitochondrial β-oxidation is the most prominent pathway for fatty acid oxidation but alternative oxidative metabolism exists. Fatty acid ω-oxidation is one of these pathways and forms dicarboxylic acids as products. These dicarboxylic acids are metabolized through peroxisomal β-oxidation representing an alternative pathway, which could potentially limit the toxic effects of fatty acid accumulation. Although dicarboxylic acid metabolism is highly active in liver and kidney, its role in physiology has not been explored in depth. In this review, we summarize the biochemical mechanism of the formation and degradation of dicarboxylic acids through ω- and β-oxidation, respectively. We will discuss the role of dicarboxylic acids in different (patho)physiological states with a particular focus on the role of the intermediates and products generated through peroxisomal β-oxidation. This review is expected to increase the understanding of dicarboxylic acid metabolism and spark future research.
Collapse
Affiliation(s)
- Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| |
Collapse
|
9
|
Zhou F, Yang L, Sun W, Wang X, Guo N, Ma H, Yang L. The PPARα/CYP4A14 bile acid pathway is associated with lipid metabolism disorders caused by low birth weight with high-fat diet. Food Nutr Res 2023; 67:8994. [PMID: 36794015 PMCID: PMC9899044 DOI: 10.29219/fnr.v67.8994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/14/2022] [Accepted: 11/04/2022] [Indexed: 01/25/2023] Open
Abstract
Purpose To investigate possible mechanisms underlying the greater susceptibility of lipid metabolism disorders in low birth weight (LBW) mice fed with high-fat diets (HFDs). Methods LBW mice model was established by using the pregnancy malnutrition method. Male pups were selected from LBW and normal-birth weight (NBW) offspring at random. After 3 weeks of weaning, all offspring mice were fed with HFD. Serum triglycerides (TGs), cholesterol (TC), low density lipoprotein (LDL-C), total bile acid (TAB), non-esterified fatty acid (NEFA), and mice fecal bile acid profiles were measured. Lipid deposition in liver sections was visualized by Oil Red O staining. The weight ratio of liver, muscle, and adiposity was calculated. Tandem mass tag (TMT) combined with LC-MS/MS was used to determine the differentially expressed proteins (DEPs) of liver tissue in two groups. Bioinformatics was used for further analysis of DEPs to screen key target proteins, and then Western Blot (WB) and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to validate the expressions of DEPs. Results LBW mice fed with HFD showed more severe lipid metabolism disorders in the childhood. In contrast to the NBW group, the serum bile acids and fecal ω-muricholic acid (ω-MCA) levels in the LBW group were significantly lower. LC-MS/MS analysis showed that downregulated proteins were associated with lipid metabolism, and further analysis found that these proteins are mainly concentrated in peroxisome proliferation-activated receptor (PPAR) and primary bile acid synthesis signaling pathways and are involved in cellular processes and metabolic processes through binding and catalytic functions. Bioinformatics analysis indicated that the level of Cytochrome P450 Family 46 Subfamily A Member 1 (CYP46A1), PPARα, key factors of cholesterol metabolism and bile acid synthesis, as well as downstream molecules Cytochrome P450 Family 4 Subfamily A Member 14 (CYP4A14), and Acyl-Coenzyme A Oxidase 2 (ACOX2) are markedly different in the liver of LBW individuals fed with HFD, and confirmed by WB and RT-qPCR. Conclusion LBW mice are more prone to dyslipidemia probably due to downregulated bile acid metabolism-related PPARα/CYP4A14 pathway, resulting in insufficient metabolism of cholesterol to bile acids, which, in turn, leads to elevated blood cholesterol.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China,Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
| | - Linquan Yang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
| | - Wenwen Sun
- Department of Internal Medicine, North China University of Science and Technology, Tangshan, China
| | - Xing Wang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
| | - Na Guo
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
| | - Huijuan Ma
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China,Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China,Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China,Huijuan Ma Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei, China.
| | - Linlin Yang
- Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China,Linlin Yang Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei, China.
| |
Collapse
|
10
|
Sun L, Xin Q, Jiao H, Wang X, Zhao J, Li H, Zhou Y, Cao A, Wang J, Lin H. Effect of exogenous bile salts supplementation on the performance and hepatic lipid metabolism of aged laying hens. J Anim Sci 2023; 101:skad334. [PMID: 37773415 PMCID: PMC11025372 DOI: 10.1093/jas/skad334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/28/2023] [Indexed: 10/01/2023] Open
Abstract
Bile acids (BA), a series of hydroxylated steroids secreted by the liver, are involved in the digestion and absorption of dietary fats. In the present study, the effect of exogenous BAs on the performance and liver lipid metabolism of laying hens was investigated. Three hundred and sixty 50-wk-old Hy-line Brown hens were randomly allocated into three groups and subjected to one of the following treatments: fed with the basal diet (control, Con), the basal diet supplemented with 0.1 g/kg (0.1 g/kg BAs), or 0.2 g/kg (0.2 g/kg BAs) porcine BAs. Laying performance, egg quality, and blood parameters were measured during the 8-wk experimental period. The expression of genes related to hepatic lipid metabolism was determined at the end of experiment. The results showed that BAs treatments had no influence (P > 0.05) on laying rate, egg weight, and feed efficiency. BAs treatment, however, significantly decreased mortality of hens (P = 0.006). BAs treatment showed a transient negative influence on eggshell quality at week 4 but not at week 8. The yolk color on week 8 was increased by BAs treatments (P < 0.0001) compared to control. The duodenum index showed a tendency to be increased (P = 0.053) and jejunum index were increased (P = 0.007) by BAs treatment. Compared to control, BAs treatments decreased lipid droplet content (P < 0.0001) and TG content (P = 0.002) of liver. Fatty acid synthase activity was also decreased as an effect of BAs dietary supplementation. Compared to the control group, 0.1 g/kg BAs treatment increased (P < 0.05) the mRNA expression of genes Farnesoid X receptor (FXR) (P = 0.042), cytochrome P450 family 7 subfamily A member 1 (CYP7A1) (P = 0.002), and cytochrome P450 family 8 subfamily B member 1 (CYP8B1) (P = 0.017), fatty acid synthase (FAS) (P = 0.020), acetyl-CoA carboxylase (ACC) (P = 0.032), sterol regulatory element binding protein-1c (SREBP-1c) (P = 0.037), proliferator-activated receptor gamma (PPARγ) (P = 0.002), apolipoprotein B (APO-B) (P = 0.020), and very low density lipoprotein receptor (VLDLR) (P = 0.024). In conclusion, the addition of exogenous BAs reduces lipid accumulation in liver. BA supplementation reduces the mortality of hens and improves egg yolk color, with no unfavorable effect on laying performance. The result suggests that suppressed FAS activity is involved in the reduced hepatic lipid accumulation by BAs treatment.
Collapse
Affiliation(s)
- Lijing Sun
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| | - Qian Xin
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| | - Hongchao Jiao
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| | - Xiaojuan Wang
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| | - Jingpeng Zhao
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| | - Haifang Li
- College of Life Sciences, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Yunlei Zhou
- College of Chemistry and Material Science, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PR China
| | - Aizhi Cao
- Shandong Longchang Animal Health Products Co., Ltd., Jingshi Street, Jinan City, Shandong Province 250000, PR China
| | - Jianmin Wang
- Shandong Longchang Animal Health Products Co., Ltd., Jingshi Street, Jinan City, Shandong Province 250000, PR China
| | - Hai Lin
- College of Animal Science and Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, 61, Daizong Street, Taian City, Shandong Province 271018, PRChina
| |
Collapse
|
11
|
Kocherlakota S, Swinkels D, Van Veldhoven PP, Baes M. Mouse Models to Study Peroxisomal Functions and Disorders: Overview, Caveats, and Recommendations. Methods Mol Biol 2023; 2643:469-500. [PMID: 36952207 DOI: 10.1007/978-1-0716-3048-8_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
During the last three decades many mouse lines were created or identified that are deficient in one or more peroxisomal functions. Different methodologies were applied to obtain global, hypomorph, cell type selective, inducible, and knockin mice. Whereas some models closely mimic pathologies in patients, others strongly deviate or no human counterpart has been reported. Often, mice, apparently endowed with a stronger transcriptional adaptation, have to be challenged with dietary additions or restrictions in order to trigger phenotypic changes. Depending on the inactivated peroxisomal protein, several approaches can be taken to validate the loss-of-function. Here, an overview is given of the available mouse models and their most important characteristics.
Collapse
Affiliation(s)
- Sai Kocherlakota
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniëlle Swinkels
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Impact of Liver Inflammation on Bile Acid Side Chain Shortening and Amidation. Cells 2022; 11:cells11243983. [PMID: 36552746 PMCID: PMC9777420 DOI: 10.3390/cells11243983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Bile acid (BA) synthesis from cholesterol by hepatocytes is inhibited by inflammatory cytokines. Whether liver inflammation also affects BA side chain shortening and conjugation was investigated. In human liver cell lines (IHH, HepG2, and HepaRG), agonists of nuclear receptors including the farnesoid X receptor (FXR), liver X receptor (LXR), and peroxisome proliferator-activated receptors (PPARs) did not affect the expression of BA-related peroxisomal enzymes. In contrast, hepatocyte nuclear factor 4α (HNF4α) inhibition down-regulated acyl-CoA oxidase 2 (ACOX2). ACOX2 was repressed by fibroblast growth factor 19 (FGF19), which was prevented by extracellular signal-regulated kinase (ERK) pathway inhibition. These changes were paralleled by altered BA synthesis (HPLC-MS/MS). Cytokines able to down-regulate cholesterol-7α-hydroxylase (CYP7A1) had little effect on peroxisomal enzymes involved in BA synthesis except for ACOX2 and bile acid-CoA:amino acid N-acyltransferase (BAAT), which were down-regulated, mainly by oncostatin M (OSM). This effect was prevented by Janus kinase (JAK) inhibition, which restored BA side chain shortening and conjugation. The binding of OSM to the extracellular matrix accounted for a persistent effect after culture medium replacement. In silico analysis of four databases (n = 201) and a validation cohort (n = 90) revealed an inverse relationship between liver inflammation and ACOX2/BAAT expression which was associated with changes in HNF4α levels. In conclusion, BA side chain shortening and conjugation are inhibited by inflammatory effectors. However, other mechanisms involved in BA homeostasis counterbalance any significant impact on the serum BA profile.
Collapse
|
14
|
Alonso‐Peña M, Espinosa‐Escudero R, Herraez E, Briz O, Cagigal ML, Gonzalez‐Santiago JM, Ortega‐Alonso A, Fernandez‐Rodriguez C, Bujanda L, Calvo Sanchez M, D´Avola D, Londoño M, Diago M, Fernandez‐Checa JC, Garcia‐Ruiz C, Andrade RJ, Lammert F, Prieto J, Crespo J, Juamperez J, Diaz‐Gonzalez A, Monte MJ, Marin JJG. Beneficial effect of ursodeoxycholic acid in patients with acyl-CoA oxidase 2 (ACOX2) deficiency-associated hypertransaminasemia. Hepatology 2022; 76:1259-1274. [PMID: 35395098 PMCID: PMC9796151 DOI: 10.1002/hep.32517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS A variant (p.Arg225Trp) of peroxisomal acyl-CoA oxidase 2 (ACOX2), involved in bile acid (BA) side-chain shortening, has been associated with unexplained persistent hypertransaminasemia and accumulation of C27-BAs, mainly 3α,7α,12α-trihydroxy-5β-cholestanoic acid (THCA). We aimed to investigate the prevalence of ACOX2 deficiency-associated hypertransaminasemia (ADAH), its response to ursodeoxycholic acid (UDCA), elucidate its pathophysiological mechanism and identify other inborn errors that could cause this alteration. METHODS AND RESULTS Among 33 patients with unexplained hypertransaminasemia from 11 hospitals and 13 of their relatives, seven individuals with abnormally high C27-BA levels (>50% of total BAs) were identified by high-performance liquid chromatography-mass spectrometry. The p.Arg225Trp variant was found in homozygosity (exon amplification/sequencing) in two patients and three family members. Two additional nonrelated patients were heterozygous carriers of different alleles: c.673C>T (p.Arg225Trp) and c.456_459del (p.Thr154fs). In patients with ADAH, impaired liver expression of ACOX2, but not ACOX3, was found (immunohistochemistry). Treatment with UDCA normalized aminotransferase levels. Incubation of HuH-7 hepatoma cells with THCA, which was efficiently taken up, but not through BA transporters, increased reactive oxygen species production (flow cytometry), endoplasmic reticulum stress biomarkers (GRP78, CHOP, and XBP1-S/XBP1-U ratio), and BAXα expression (reverse transcription followed by quantitative polymerase chain reaction and immunoblot), whereas cell viability was decreased (tetrazolium salt-based cell viability test). THCA-induced cell toxicity was higher than that of major C24-BAs and was not prevented by UDCA. Fourteen predicted ACOX2 variants were generated (site-directed mutagenesis) and expressed in HuH-7 cells. Functional tests to determine their ability to metabolize THCA identified six with the potential to cause ADAH. CONCLUSIONS Dysfunctional ACOX2 has been found in several patients with unexplained hypertransaminasemia. This condition can be accurately identified by a noninvasive diagnostic strategy based on plasma BA profiling and ACOX2 sequencing. Moreover, UDCA treatment can efficiently attenuate liver damage in these patients.
Collapse
Affiliation(s)
- Marta Alonso‐Peña
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive DiseasesValdecilla Research Institute (IDIVAL)Marqués de Valdecilla University HospitalSantanderSpain
| | - Ricardo Espinosa‐Escudero
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
| | - Elisa Herraez
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
| | - Oscar Briz
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
| | - Maria Luisa Cagigal
- Pathological Anatomy ServiceHospital Universitario Marqués de ValdecillaSantanderSpain
| | - Jesus M. Gonzalez‐Santiago
- Department of Gastroenterology and HepatologyUniversity Hospital of SalamancaInstitute for Biomedical ResearchSalamancaSpain
| | - Aida Ortega‐Alonso
- Liver Unit, Gastroenterology ServiceInstitute of Biomedical Research of MálagaSchool of MedicineUniversity Hospital Virgen de la VictoriaMálagaSpain
| | | | - Luis Bujanda
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Department of Liver and Gastrointestinal DiseasesBiodonostia Health Research InstituteDonostia University HospitalUniversity of the Basque CountrySan SebastianSpain
| | | | - Delia D´Avola
- Department of MedicineClinica Universidad de Navarra and Center for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Maria‐Carlota Londoño
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Liver UnitHospital Clínic de BarcelonaUniversity of BarcelonaBarcelonaSpain
- Institute of Biomedical Research of Barcelona (IDIBAPS)BarcelonaSpain
| | - Moises Diago
- Valencia University General HospitalValenciaSpain
| | - Jose C. Fernandez‐Checa
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Institute of Biomedical Research of Barcelona (IDIBAPS)BarcelonaSpain
- Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Research Center for Alcoholic Liver and Pancreatic Diseases (ALPD) and CirrhosisKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Carmen Garcia‐Ruiz
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Institute of Biomedical Research of Barcelona (IDIBAPS)BarcelonaSpain
- Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Research Center for Alcoholic Liver and Pancreatic Diseases (ALPD) and CirrhosisKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Raul J. Andrade
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Liver Unit, Gastroenterology ServiceInstitute of Biomedical Research of MálagaSchool of MedicineUniversity Hospital Virgen de la VictoriaMálagaSpain
| | - Frank Lammert
- Department of Medicine IISaarland University Medical CenterHomburgGermany
- Health SciencesHannover Medical SchoolHannoverGermany
| | - Jesus Prieto
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
- Department of MedicineClinica Universidad de Navarra and Center for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive DiseasesValdecilla Research Institute (IDIVAL)Marqués de Valdecilla University HospitalSantanderSpain
| | - Javier Juamperez
- Pediatric Hepatology and Liver Transplantation UnitVall d’Hebron University HospitalUniversitat Autónoma de BarcelonaBarcelonaSpain
| | - Alvaro Diaz‐Gonzalez
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive DiseasesValdecilla Research Institute (IDIVAL)Marqués de Valdecilla University HospitalSantanderSpain
| | - Maria J. Monte
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
| | - Jose J. G. Marin
- Experimental Hepatology and Drug TargetingInstitute for Biomedical ResearchUniversity of SalamancaSalamancaSpain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD)Carlos III National Institute of HealthMadridSpain
| |
Collapse
|
15
|
Zhang Y, Chen Y, Zhang Z, Tao X, Xu S, Zhang X, Zurashvili T, Lu Z, Bayascas JR, Jin L, Zhao J, Zhou X. Acox2 is a regulator of lysine crotonylation that mediates hepatic metabolic homeostasis in mice. Cell Death Dis 2022; 13:279. [PMID: 35351852 PMCID: PMC8964741 DOI: 10.1038/s41419-022-04725-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/22/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022]
Abstract
Acyl-CoA oxidase 2 (Acox2) is an enzyme involved in peroxisomal bile acid synthesis and branched-chain fatty acid degradation. Acox2 knockout (−/−) mice spontaneously developed liver cancer with marked lymphocytic infiltrate. Tandem-affinity purification coupled with mass spectrometry analysis revealed that Acox2 interacted with methylcrotonoyl-CoA carboxylase followed by co-immunoprecipitation confirmation. Here we reported that non-histone lysine crotonylation (Kcr) levels were downregulated in Acox2−/− mice livers. Interestingly, Kcr signals were concentrated in the nucleus of tumor cells but mostly located in the cytoplasm of adjacent normal liver cells of Acox2−/− mice. Quantitative analysis of the global crotonylome further revealed that 54% (27/50) of downregulated non-histone Kcr sites were located in mitochondrial (11/50) and peroxisomal (17/50) enzymes including Ehhadh, Scp2, Hsd17b4, Crot, Etfa, Cpt1a, Eci1/2, Hadha, Etfdh, and Idh2. Subsequent site-directed mutagenesis and transcriptome analysis revealed that Ehhadh K572cr might have site-specific regulatory roles by downregulating TOP3B expression that lead to increased DNA damage in vitro. Our findings suggested Acox2 is a regulator of Kcr that might play critical role on hepatic metabolic homeostasis.
Collapse
|
16
|
Li R, Guo C, Lin X, Chan TF, Su M, Zhang Z, Lai KP. Integrative omics analysis reveals the protective role of vitamin C on perfluorooctanoic acid-induced hepatoxicity. J Adv Res 2022; 35:279-294. [PMID: 35024202 PMCID: PMC8721266 DOI: 10.1016/j.jare.2021.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 01/09/2023] Open
Abstract
Introduction Perfluorooctanoic acid (PFOA) is a compound used as an industrial surfactant in chemical processes worldwide. Population and cross-sectional studies have demonstrated positive correlations between PFOA levels and human health problems. Objectives Many studies have focused on the hepatotoxicity and liver problems caused by PFOA, with little attention to remediation of these problems. As an antioxidant, vitamin C is frequently utilized as a supplement for hepatic detoxification. Methods In this study, we use a mouse model to study the possible role of vitamin C in reducing PFOA-induced liver damage. Based on comparative transcriptomic and metabolomic analysis, we elucidate the mechanisms underlying the protective effect of vitamin C. Results Our results show that vitamin C supplementation reduces signs of PFOA-induced liver damage including total cholesterol and triglyceride levels increase, liver damage markers aspartate, transaminase, and alanine aminotransferase elevation, and liver enlargement. Further, we show that the protective role of vitamin C is associated with signaling networks control, suppressing linoleic acid metabolism, reducing thiodiglycolic acid, and elevating glutathione in the liver. Conclusion The findings in this study demonstrate, for the first time, the utility of vitamin C for preventing PFOA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Chao Guo
- Department of Pharmacy, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, PR China
| | - Xiao Lin
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ting Fung Chan
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | | | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| |
Collapse
|
17
|
Jiang J, Liu Y, Yang S, Peng H, Liu J, Cheng YX, Li N. Photoaffinity-Based Chemical Proteomics Reveals 7-Oxocallitrisic Acid Targets CPT1A to Trigger Lipogenesis Inhibition. ACS Med Chem Lett 2021; 12:1905-1911. [PMID: 34917253 DOI: 10.1021/acsmedchemlett.1c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/28/2021] [Indexed: 11/29/2022] Open
Abstract
One of the natural terpenoids isolated from Resina Commiphora, 7-oxocallitrisic acid (7-OCA), has lipid metabolism regulatory activity. To uncover its lipogenesis inhibition mechanism, we developed a photoaffinity and clickable probe based on the 7-OCA scaffold and performed chemical proteomics to profile its potential cellular targets. It was found that 7-OCA could directly interact with carnitine palmitoyl transferase 1A (CPT1A) to promote its activity to reduce lipid accumulation. The present work reveals our understanding of the mode of lipid mebabolism regulation by abietic acids and provides new clues for antiobesity drug development with CPT1A as a main target.
Collapse
Affiliation(s)
- Jianbing Jiang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Ying Liu
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Shuxin Yang
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huipai Peng
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawang Liu
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Yong-Xian Cheng
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Nan Li
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
18
|
Lyu J, Li H, Yin D, Zhao M, Sun Q, Guo M. Analysis of eight bile acids in urine of gastric cancer patients based on covalent organic framework enrichment coupled with liquid chromatography-tandem mass spectrometry. J Chromatogr A 2021; 1653:462422. [PMID: 34348207 DOI: 10.1016/j.chroma.2021.462422] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Gastric carcinoma is one of the most common and deadly forms of cancer. Early detection is critical for successful treatment of gastric cancer, and examination of BAs in urine may provide a critical diagnostic tool for identifying gastric cancer at stages when it can still be cured. Bile acids (BAs) are a crucial toxic factor correlated with the injury of gastric mucosa and as such, quantifying the amount of BA in patient's urine could provide a new means to quickly and non-invasively identify the presence of gastric cancer in the early stages. Here, a covalent organic framework (COF) material synthesized on the basis of 1,3,5-tris(4-nitrophenyl)benzene (TAPB) and pyromellitic dianhydride (PMDA) was used as stationary phase for SPE column that was coupled to LC-MS/MS for quantitative analysis of eight BAs in human urine, including cholic acid (CA), deoxycholic acid (DCA), glycochenodeoxycholic acid (GCDCA), glycocholic acid (GCA), taurochenodeoxycholic acid (TCDCA), lithocholic acid (LCA), hyodeoxycholic acid (HDCA), and chenodeoxycholic acid (CDCA). The enrichment effect of synthesized COF material was better than commercial SPE and HLB column. The sensitivity can increase 9.37- to 54.30- fold (calculated by the ratio of peak area between before and after enrichment). The probable mechanism is due to the great porosity and the similar polarity with BAs of the COF material. By compared with previous literatures, our method had the minimum limit of detection, which achieved 46.40, 25.75, 47.40, 47.37, 30.42, and 33.92 pg /mL, respectively, for GCA, GCDCA, CA, CDCA, HDCA and DCA after enrichment. These eight BAs also accomplished excellent linearity from 0.34 to 10,000 ng/mL. This material was successfully applied in the measurements of these six BAs in human urine from 76 gastric cancer patients and 32 healthy people. Compared to healthy people, levels of CA, CDCA, DCA, and HDCA were significantly elevated and levels of GCDCA were depressed, respectively, in gastric cancer patients. Our work suggests that these acids may act as potential biomarkers for gastric cancer and our framework provides a method for "non-invasive" diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Jinxiu Lyu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Haijuan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Dengyang Yin
- Jingjiang People's Hospital, Taizhou, Jiangsu, 214500, China
| | - Meng Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Qiang Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Mengzhe Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
19
|
Early Heat Exposure Effects on Proteomic Changes of the Broiler Liver under Acute Heat Stress. Animals (Basel) 2021; 11:ani11051338. [PMID: 34066761 PMCID: PMC8151403 DOI: 10.3390/ani11051338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Early heat exposure have been studied in the poultry industry as a method of reducing heat stress (HS) on poultry. However, the results of each study are inconsistent, and it has not been confirmed which mechanisms reduce HS by early heat exposure. Therefore, we tried to confirm the relaxation mechanism through proteomic analysis after applying early and acute heat exposure to broilers. The broilers were divided into three treatments, followed by CC (control group), CH (acute HS at the 35th day), and HH (early heat exposure at the fifth day and acute HS at the 35th day. Liver samples were collected and analyzed for proteomics and functional analysis. Proteins related to various functions, such as carbohydrate metabolism, fatty acid metabolism, energy metabolism, and the oxidation–reduction process, which were dramatically changed by acute HS, and were alleviated similar to the control group by early heat exposure. Through these results, the mechanism by which early heat exposure induces homeostasis during acute HS, and the possibility of the early heat exposure as a method of reducing HS were confirmed. Abstract As environmental temperatures continue to rise, heat stress (HS) is having a negative effect on the livestock industry. In order to solve this problem, many studies have been conducted to reduce HS. Among them, early heat exposure has been suggested as a method for reducing HS in poultry. In this study, we analyzed proteomics and tried to identify the metabolic mechanisms of early heat exposure on acute HS. A total of 48 chicks were separated into three groups: CC (control groups raised at optimum temperature), CH (raised with CC but exposed acute HS at the 35th day), and HH (raised with CC but exposed early heat at the fifth day and acute HS at the 35th day). After the whole period, liver samples were collected for proteomic analysis. A total of 97 differentially expressed proteins were identified by acute HS. Of these, 62 proteins recovered their expression levels by early heat exposure. We used these 62 proteins to determine the protective effects of early heat exposure. Of the various protein-related terms, we focused on the oxidative phosphorylation, fatty acid metabolism, carbohydrate metabolism, and energy production metabolism. Our findings suggest the possibility of early heat exposure effects in acute HS that may be useful in breeding or management techniques for producing broilers with high heat resistance.
Collapse
|
20
|
Zhang Y, Lu Z, Zeng W, Zhao J, Zhou X. Two sides of NNMT in alcoholic and non-alcoholic fatty liver development. J Hepatol 2021; 74:1250-1253. [PMID: 33340570 DOI: 10.1016/j.jhep.2020.11.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 12/04/2022]
Affiliation(s)
- Yuan Zhang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Zhouping Lu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Weijia Zeng
- State Key Lab of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jianyuan Zhao
- State Key Lab of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China; Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiangyu Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China; Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.
| |
Collapse
|
21
|
Das Y, Swinkels D, Baes M. Peroxisomal Disorders and Their Mouse Models Point to Essential Roles of Peroxisomes for Retinal Integrity. Int J Mol Sci 2021; 22:ijms22084101. [PMID: 33921065 PMCID: PMC8071455 DOI: 10.3390/ijms22084101] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/31/2022] Open
Abstract
Peroxisomes are multifunctional organelles, well known for their role in cellular lipid homeostasis. Their importance is highlighted by the life-threatening diseases caused by peroxisomal dysfunction. Importantly, most patients suffering from peroxisomal biogenesis disorders, even those with a milder disease course, present with a number of ocular symptoms, including retinopathy. Patients with a selective defect in either peroxisomal α- or β-oxidation or ether lipid synthesis also suffer from vision problems. In this review, we thoroughly discuss the ophthalmological pathology in peroxisomal disorder patients and, where possible, the corresponding animal models, with a special emphasis on the retina. In addition, we attempt to link the observed retinal phenotype to the underlying biochemical alterations. It appears that the retinal pathology is highly variable and the lack of histopathological descriptions in patients hampers the translation of the findings in the mouse models. Furthermore, it becomes clear that there are still large gaps in the current knowledge on the contribution of the different metabolic disturbances to the retinopathy, but branched chain fatty acid accumulation and impaired retinal PUFA homeostasis are likely important factors.
Collapse
|
22
|
Zhang Q, Zhang Y, Sun S, Wang K, Qian J, Cui Z, Tao T, Zhou J. ACOX2 is a prognostic marker and impedes the progression of hepatocellular carcinoma via PPARα pathway. Cell Death Dis 2021; 12:15. [PMID: 33414412 PMCID: PMC7791021 DOI: 10.1038/s41419-020-03291-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) has been extensively studied as one of the most aggressive tumors worldwide. However, its mortality rate remains high due to ideal diagnosis and treatment strategies. Uncovering novel genes with prognostic significance would shed light on improving the HCC patient's outcome. In our study, we applied data-independent acquisition (DIA) quantitative proteomics to investigate the expression landscape of 24 paired HCC patients. A total of 1029 differentially expressed proteins (DEPs) were screened. Then, we compared DEPs in our cohort with the differentially expressed genes (DEGs) in The Cancer Genome Atlas, and investigated their prognostic significance, and found 183 prognosis-related genes (PRGs). By conducting protein-protein interaction topological analysis, we identified four subnetworks with prognostic significance. Acyl-CoA oxidase 2 (ACOX2) is a novel gene in subnetwork1, encodes a peroxisomal enzyme, and its function in HCC was investigated in vivo and in vitro. The lower expression of ACOX2 was validated by real-time quantitative PCR, immunohistochemistry, and Western blot. Cell Counting Kit-8 assay, wound healing, and transwell migration assay were applied to evaluate the impact of ACOX2 overexpression on the proliferation and migration abilities in two liver cancer cell lines. ACOX2 overexpression, using a subcutaneous xenograft tumor model, indicated a tumor suppressor role in HCC. To uncover the underlying mechanism, gene set enrichment analysis was conducted, and peroxisome proliferator-activated receptor-α (PPARα) was proposed to be a potential target. In conclusion, we demonstrated a PRG ACOX2, and its overexpression reduced the proliferation and metastasis of liver cancer in vitro and in vivo through PPARα pathway.
Collapse
Affiliation(s)
- Qifan Zhang
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Yunbin Zhang
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shibo Sun
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Kai Wang
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Jianping Qian
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Zhonglin Cui
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Tao Tao
- Department of Anesthesiology, Central People's Hospital of Zhanjiang, 236 Yuanzhu Road, Zhanjiang, Guangdong, 524045, China.
| | - Jie Zhou
- Department of General Surgery, Division of Hepatobiliopancreatic Surgery, Nanfang Hospital, Southern Medical University, 1838 North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
23
|
Kim JT, Won SY, Kang K, Kim SH, Park MS, Choi KH, Nam TS, Denis SW, Ferdinandusse S, Lee JE, Choi SY, Kim MK. ACOX3 Dysfunction as a Potential Cause of Recurrent Spontaneous Vasospasm of Internal Carotid Artery. Transl Stroke Res 2020; 11:1041-1051. [PMID: 31975215 DOI: 10.1007/s12975-020-00779-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 11/24/2022]
Abstract
Recurrent spontaneous vasospasm of the extracranial internal carotid artery (RSV-eICA) is a rarely recognized cause of ischemic stroke in young adults. However, its pathophysiology remains largely unknown. Through whole-exome sequencing of the ACOX3 gene of two dizygotic Korean twin brothers affected by RSV-eICA, we identified two compound heterozygous missense variants c.235 T > G (p.F79 V) and c.665G > A (p.G222E). In silico analysis indicated that both variants were classified as pathogenic. In vitro ACOX3 enzyme assay indicated practically no enzyme activity in both F79 V and G222E mutants. To determine the effect of the mutants on vasospasm, we used a collagen contraction assay on human aortic smooth muscle cells (HASMC). Carbachol, a cholinergic agonist, induces contraction of HASMC. Knockdown of ACOX3 in HASMC, using siRNA, significantly repressed HASMC contraction triggered by carbachol. The carbachol-induced HASMC contraction was restored by transfection with plasmids encoding siRNA-resistant wild-type ACOX3, but not by transfection with ACOX3 G222E or by co-transfection with ACOX3 F79 V and ACOX3 G222E, indicating that the two ACOX3 mutants suppress carbachol-induced HASMC contraction. We propose that an ACOX3 dysfunction elicits a prolonged loss of the basal aortic myogenic tone. As a result, smooth muscles of the ICA's intermediate segment, in which the sympathetic innervation is especially rich, becomes hypersensitive to sympathomimetic stimuli (e.g., heavy exercise) leading to a recurrent vasospasm. Therefore, ACOX3 dysfunction would be a causal mechanism of RSV-eICA. For the first time, we report the possible involvement of ACOX3 in maintaining the basal myogenic tone of human arterial smooth muscle.
Collapse
Affiliation(s)
- Joon-Tae Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - So Yeon Won
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea
| | - KyungWook Kang
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Sang-Hoon Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Man-Seok Park
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Kang-Ho Choi
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea
| | - Simone W Denis
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Academic Medical Center, Amsterdam, The Netherlands
| | - Ji Eun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, South Korea.
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| | - Myeong-Kyu Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, 61469, South Korea.
| |
Collapse
|
24
|
Wanders RJA, Vaz FM, Waterham HR, Ferdinandusse S. Fatty Acid Oxidation in Peroxisomes: Enzymology, Metabolic Crosstalk with Other Organelles and Peroxisomal Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1299:55-70. [PMID: 33417207 DOI: 10.1007/978-3-030-60204-8_5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Peroxisomes play a central role in metabolism as exemplified by the fact that many genetic disorders in humans have been identified through the years in which there is an impairment in one or more of these peroxisomal functions, in most cases associated with severe clinical signs and symptoms. One of the key functions of peroxisomes is the β-oxidation of fatty acids which differs from the oxidation of fatty acids in mitochondria in many respects which includes the different substrate specificities of the two organelles. Whereas mitochondria are the main site of oxidation of medium-and long-chain fatty acids, peroxisomes catalyse the β-oxidation of a distinct set of fatty acids, including very-long-chain fatty acids, pristanic acid and the bile acid intermediates di- and trihydroxycholestanoic acid. Peroxisomes require the functional alliance with multiple subcellular organelles to fulfil their role in metabolism. Indeed, peroxisomes require the functional interaction with lysosomes, lipid droplets and the endoplasmic reticulum, since these organelles provide the substrates oxidized in peroxisomes. On the other hand, since peroxisomes lack a citric acid cycle as well as respiratory chain, oxidation of the end-products of peroxisomal fatty acid oxidation notably acetyl-CoA, and different medium-chain acyl-CoAs, to CO2 and H2O can only occur in mitochondria. The same is true for the reoxidation of NADH back to NAD+. There is increasing evidence that these interactions between organelles are mediated by tethering proteins which bring organelles together in order to allow effective exchange of metabolites. It is the purpose of this review to describe the current state of knowledge about the role of peroxisomes in fatty acid oxidation, the transport of metabolites across the peroxisomal membrane, its functional interaction with other subcellular organelles and the disorders of peroxisomal fatty acid β-oxidation identified so far in humans.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases and Emma Children's hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Frédéric M Vaz
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases and Emma Children's hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans R Waterham
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases and Emma Children's hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Ferdinandusse
- Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases and Emma Children's hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Xu W, Liu Z, Ren H, Peng X, Wu A, Ma D, Liu G, Liu L. Twenty Metabolic Genes Based Signature Predicts Survival of Glioma Patients. J Cancer 2020; 11:441-449. [PMID: 31897239 PMCID: PMC6930419 DOI: 10.7150/jca.30923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Glioma, caused by carcinogenesis of brain and spinal glial cells, is the most common primary malignant brain tumor. To find the important indicator for glioma prognosis is still a challenge and the metabolic alteration of glioma has been frequently reported recently. Methods: In our current work, a risk score model based on the expression of twenty metabolic genes was developed using the metabolic gene expressions in The Cancer Genome Atlas (TCGA) dataset, the methods of which included the cox multivariate regression and the random forest variable hunting, a kind of machine learning algorithm, and the risk score generated from this model is used to make predictions in the survival of glioma patients in the training dataset. Subsequently, the result was further verified in other three verification sets (GSE4271, GSE4412 and GSE16011). Risk score related pathways collected in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database were identified using Gene Set Enrichment Analysis (GSEA). Results: The risk score generated from our model makes good predictions in the survival of glioma patients in the training dataset and other three verification sets. By assessing the relationships between clinical indicators and the risk score, we found that the risk score was an independent and significant indicator for the prognosis of glioma patients. Simultaneously, we conducted a survival analysis of the patients who received chemotherapy and who did not, finding that the risk score was equally valid in both cases. And signaling pathways related to the genesis and development of multiple cancers were also identified. Conclusions: In summary, our risk score model is predictive for 967 glioma patients' survival from four independent datasets, and the risk score is a meaningful and independent parameter of the clinicopathological information.
Collapse
Affiliation(s)
- Wenfang Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Zhenhao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - He Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Xueqing Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Aoshen Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Duan Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Gang Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| | - Lei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R.China
| |
Collapse
|
26
|
Crick PJ, Yutuc E, Abdel-Khalik J, Saeed A, Betsholtz C, Genove G, Björkhem I, Wang Y, Griffiths WJ. Formation and metabolism of oxysterols and cholestenoic acids found in the mouse circulation: Lessons learnt from deuterium-enrichment experiments and the CYP46A1 transgenic mouse. J Steroid Biochem Mol Biol 2019; 195:105475. [PMID: 31541728 PMCID: PMC6880786 DOI: 10.1016/j.jsbmb.2019.105475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/31/2022]
Abstract
While the presence and abundance of the major oxysterols and cholestenoic acids in the circulation is well established, minor cholesterol metabolites may also have biological importance and be of value to investigate. In this study by observing the metabolism of deuterium-labelled cholesterol in the pdgfbret/ret mouse, a mouse model with increased vascular permeability in brain, and by studying the sterol content of plasma from the CYP46A1 transgenic mouse overexpressing the human cholesterol 24S-hydroxylase enzyme we have been able to identify a number of minor cholesterol metabolites found in the circulation, make approximate-quantitative measurements and postulate pathways for their formation. These "proof of principle" data may have relevance when using mouse models to mimic human disease and in respect of the increasing possibility of treating human neurodegenerative diseases with pharmaceuticals designed to enhance the activity of CYP46A1 or by adeno-associated virus delivery of CYP46A1.
Collapse
Affiliation(s)
- Peter J Crick
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK
| | - Eylan Yutuc
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK
| | - Jonas Abdel-Khalik
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK
| | - Ahmed Saeed
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital, Karolinska Institutet, 141 86 Huddinge, Sweden
| | | | - Guillem Genove
- ICMC Karolinska Institutet, Novum, 141 57 Huddinge, Sweden
| | - Ingemar Björkhem
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital, Karolinska Institutet, 141 86 Huddinge, Sweden
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK.
| | - William J Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK.
| |
Collapse
|
27
|
Abstract
In mammalian systems "sterolomics" can be regarded as the quantitative or semi-quantitative profiling of all metabolites derived from cholesterol and its cyclic precursors. The system can be further complicated by metabolites derived from ingested phytosterols or pharmaceuticals, but this is beyond the scope of this article. "Sterolomics" can be performed on either an unbiased global format, or more usually, exploiting a targeted format. Here we discuss the different mass spectrometry-based analytical techniques used in "sterolomics" giving specific examples in the context of neurodegenerative disease and for the diagnosis of inborn errors of metabolism. We pay particular attention to the profiling of cholesterol metabolites in the bile acid biosynthesis pathways, although the analytical techniques discussed are also appropriate for analysis of hormonal steroids.
Collapse
Affiliation(s)
- William J. Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea SA2 8PP, Wales, UK
| |
Collapse
|
28
|
Oxysterol research: a brief review. Biochem Soc Trans 2019; 47:517-526. [PMID: 30936243 PMCID: PMC6490702 DOI: 10.1042/bst20180135] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/16/2022]
Abstract
In the present study, we discuss the recent developments in oxysterol research. Exciting results have been reported relating to the involvement of oxysterols in the fields of neurodegenerative disease, especially in Huntington's disease, Parkinson's disease and Alzheimer's disease; in signalling and development, in particular, in relation to Hedgehog signalling; and in cancer, with a special focus on (25R)26-hydroxycholesterol. Methods for the measurement of oxysterols, essential for understanding their mechanism of action in vivo, and valuable for diagnosing rare diseases of cholesterol biosynthesis and metabolism are briefly considered.
Collapse
|
29
|
Sharma V, Hiller M. Loss of Enzymes in the Bile Acid Synthesis Pathway Explains Differences in Bile Composition among Mammals. Genome Biol Evol 2018; 10:3211-3217. [PMID: 30388264 PMCID: PMC6296402 DOI: 10.1093/gbe/evy243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2018] [Indexed: 12/11/2022] Open
Abstract
Bile acids are important for absorbing nutrients. Most mammals produce cholic and chenodeoxycholic bile acids. Here, we investigated genes in the bile acid synthesis pathway in four mammals that deviate from the usual mammalian bile composition. First, we show that naked-mole rats, elephants, and manatees repeatedly inactivated CYP8B1, an enzyme uniquely required for cholic acid synthesis, which explains the absence of cholic acid in these species. Second, no gene-inactivating mutations were found in any pathway gene in the rhinoceros, a species that lacks bile acids, indicating an evolutionarily recent change in its bile composition. Third, elephants and/or manatees that also lack bile acids altogether have lost additional nonessential enzymes (SLC27A5, ACOX2). Apart from uncovering genomic differences explaining deviations in bile composition, our analysis of bile acid enzymes in bile acid-lacking species suggests that essentiality prevents gene loss, while loss of pleiotropic genes is permitted if their other functions are compensated by functionally related proteins.
Collapse
Affiliation(s)
- Virag Sharma
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany.,CRTD-DFG Center for Regenerative Therapies Dresden, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden; and German Center for Diabetes Research (DZD), Munich, Neuherberg, Germany
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.,Center for Systems Biology Dresden, Dresden, Germany
| |
Collapse
|
30
|
Wanders RJA, Vaz FM, Ferdinandusse S, Kemp S, Ebberink MS, Waterham HR. Laboratory Diagnosis of Peroxisomal Disorders in the -Omics Era and the Continued Importance of Biomarkers and Biochemical Studies. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2018. [DOI: 10.1177/2326409818810285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Ronald J. A. Wanders
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frédéric M. Vaz
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Merel S. Ebberink
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Hans R. Waterham
- Laboratory Genetic Metabolic Diseases, Departments of Clinical Chemistry and Pediatrics, EmmaChildren’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
31
|
Abstract
Peroxisomes are key metabolic organelles, which contribute to cellular lipid metabolism, e.g. the β-oxidation of fatty acids and the synthesis of myelin sheath lipids, as well as cellular redox balance. Peroxisomal dysfunction has been linked to severe metabolic disorders in man, but peroxisomes are now also recognized as protective organelles with a wider significance in human health and potential impact on a large number of globally important human diseases such as neurodegeneration, obesity, cancer, and age-related disorders. Therefore, the interest in peroxisomes and their physiological functions has significantly increased in recent years. In this review, we intend to highlight recent discoveries, advancements and trends in peroxisome research, and present an update as well as a continuation of two former review articles addressing the unsolved mysteries of this astonishing organelle. We summarize novel findings on the biological functions of peroxisomes, their biogenesis, formation, membrane dynamics and division, as well as on peroxisome-organelle contacts and cooperation. Furthermore, novel peroxisomal proteins and machineries at the peroxisomal membrane are discussed. Finally, we address recent findings on the role of peroxisomes in the brain, in neurological disorders, and in the development of cancer.
Collapse
Affiliation(s)
- Markus Islinger
- Institute of Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, Medical Faculty Manheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Alfred Voelkl
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | - H Dariush Fahimi
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | | |
Collapse
|
32
|
Ashby K, Navarro Almario EE, Tong W, Borlak J, Mehta R, Chen M. Review article: therapeutic bile acids and the risks for hepatotoxicity. Aliment Pharmacol Ther 2018; 47:1623-1638. [PMID: 29701277 DOI: 10.1111/apt.14678] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/17/2018] [Accepted: 03/31/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bile acids play important roles in cholesterol metabolism and signal through farnesoid X receptor and G protein-coupled receptors. Given their importance in liver biology, bile acid therapy enables therapeutic applications beyond the treatment of cholestatic liver disease. However, predicting hepatotoxicity of bile acids in humans is obscured due to inconsistent extrapolations of animal data to humans. AIM To review the evidence that could explain discordant bile acids hepatotoxicity observed in humans and animals. METHOD Literature search was conducted in PubMed using keywords "bile acid," "transporter," "hepatotoxicity," "clinical study," "animal study," "species difference," "mechanism," "genetic disorder." Relevant articles were selected for review. RESULTS Clinically significant hepatotoxicity was reported in response to certain bile acids, namely chenodeoxycholic acid, which was given a boxed warning for potential hepatotoxicity. The chemical structure, specifically the number and orientation of hydroxyl groups, significantly affects their hydrophobicity, an important factor in bile acid toxicity. Experimental studies show that hydrophobic bile acids can lead to liver injury through various mechanisms, such as death receptor signalling, mitochondrial dysfunction and inflammation. Although animal studies play a central role in investigating bile acid safety, there are considerable differences in bile acid composition, metabolism and hepatobiliary disposition across species. This does not allow appropriate safety inference, especially for predicting hepatotoxicity in humans. Exploring evidences stemming from inborn errors, genetic models of disease and toxicology studies further improves an understanding of bile acid hepatotoxicity. CONCLUSION Species differences should be considered in the development of bile acid related therapeutics. Although the mechanism of bile acid hepatotoxicity is still not fully understood, continued mechanistic studies will deepen our understanding.
Collapse
Affiliation(s)
- K Ashby
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - E E Navarro Almario
- Office of Computational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - W Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - J Borlak
- Hannover Medical School, Center of Pharmacology and Toxicology, Hannover, Germany
| | - R Mehta
- Division of Gastroenterology and Inborn Error Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - M Chen
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
33
|
Kong M, Liu HH, Wu J, Shen MQ, Wang ZG, Duan SM, Zhang YB, Zhu H, Li SL. Effects of sulfur-fumigation on the pharmacokinetics, metabolites and analgesic activity of Radix Paeoniae Alba. JOURNAL OF ETHNOPHARMACOLOGY 2018; 212:95-105. [PMID: 29080828 DOI: 10.1016/j.jep.2017.10.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/22/2017] [Accepted: 10/23/2017] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Paeoniae Alba (Baishao, BS), one of the most commonly used traditional Chinese medicinal herbs, has many pharmacological effects including analgesic activity. Previous studies found that sulfur-fumigation, a post-harvest handling process developed to prevent mold contamination of medicinal herbs, altered the quality of BS. However, whether sulfur-fumigation affects the pharmacokinetics, safety and efficacy of BS warrants further investigation. AIM OF THE STUDY To evaluate the feasibility of sulfur-fumigation as a post-harvest handling process of BS from the viewpoints of pharmacokinetics, safety and efficacy. MATERIALS AND METHODS The pharmacokinetic behaviors of four active components of BS and one characteristic component of sulfur-fumigated BS (S-BS) were evaluated by high performance liquid chromatography triple quadrupole mass spectrometry (HPLC-TQ-MS/MS). The safety was investigated using ultra high performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC-QTOF-MS/MS) based metabolomics approach after intragastric (i.g.) administration of non-fumigated BS (N-BS) and S-BS in rats. The analgesic efficacy was compared using hot-plate test in mice, after i.g. administration of N-BS and S-BS, at both high and low dosages. RESULTS Systemic exposures of paeoniflorin and oxypaeoniflorin, two analgesic components of BS, were significantly decreased in the S-BS treated group compared to the N-BS treated group, while paeoniflorin sulfonate, one of the sulfur-containing derivatives of S-BS, was detected in all time-points of S-BS treated group with the area under the plasma concentration-time curve (AUC0-t) and the maximum plasma concentration (Cmax) as high as 7077.06 ± 2232.97ng/mL*h and 1641.42 ± 634.79ng/mL respectively, which indicated that sulfur-fumigation altered the pharmacokinetic behaviors of BS. Besides, paeoniflorin sulfonate and its four metabolites with ambiguous toxicities, as well as one endogenous metabolite p-cresol glucuronide, the biomarker of disordered homeostasis of intestinal bacteria and bile acid, were identified as the characteristic metabolites in S-BS administered rats, suggesting that sulfur-fumigation reduced the safety of BS. Furthermore, the analgesic effects at both low and high dosages were decreased in different extent when compared to N-BS administered groups, indicating that sulfur-fumigation weakened the efficacy of BS. CONCLUSION Sulfur-fumigation altered the pharmacokinetics, as well as reduced the safety and efficacy of BS, suggesting that sulfur-fumigation is not recommended for post-harvest handling of BS.
Collapse
Affiliation(s)
- Ming Kong
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Huan-Huan Liu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Jie Wu
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, PR China
| | - Ming-Qin Shen
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Zhi-Gang Wang
- Department of Pharmacology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Su-Min Duan
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China
| | - Yan-Bo Zhang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - He Zhu
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, PR China.
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, PR China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine and Jiangsu Branch of China Academy of Chinese Medical Sciences, Nanjing 210028, PR China.
| |
Collapse
|
34
|
Wanders RJA. Peroxisomal disorders: Improved laboratory diagnosis, new defects and the complicated route to treatment. Mol Cell Probes 2018; 40:60-69. [PMID: 29438773 DOI: 10.1016/j.mcp.2018.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 12/15/2022]
Abstract
Peroxisomes catalyze a number of essential metabolic functions of which fatty acid alpha- and beta-oxidation, ether phospholipid biosynthesis, glyoxylate detoxification and bile acid synthesis are the most important. The key role of peroxisomes in humans is exemplified by the existence of a group of peroxisomal disorders, caused by mutations in > 30 different genes which code for proteins with a role in either peroxisome biogenesis or one of the metabolic pathways in peroxisomes. Technological advances in laboratory methods at the metabolite-, enzyme-, and molecular level have not only allowed the identification of new peroxisomal disorders but also new phenotypes associated with already identified genetic defects thus extending the clinical spectrum. Unfortunately, progress in the field of pathogenesis and treatment has lagged behind although there are certainly new and hopeful developments with respect to X-linked adrenoleukodystrophy and hyperoxaluria type 1.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands; Department of Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Ferdinandusse S, Denis S, van Roermund CWT, Preece MA, Koster J, Ebberink MS, Waterham HR, Wanders RJA. A novel case of ACOX2 deficiency leads to recognition of a third human peroxisomal acyl-CoA oxidase. Biochim Biophys Acta Mol Basis Dis 2017; 1864:952-958. [PMID: 29287774 DOI: 10.1016/j.bbadis.2017.12.032] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/12/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022]
Abstract
Peroxisomal acyl-CoA oxidases catalyze the first step of beta-oxidation of a variety of substrates broken down in the peroxisome. These include the CoA-esters of very long-chain fatty acids, branched-chain fatty acids and the C27-bile acid intermediates. In rat, three peroxisomal acyl-CoA oxidases with different substrate specificities are known, whereas in humans it is believed that only two peroxisomal acyl-CoA oxidases are expressed under normal circumstances. Only three patients with ACOX2 deficiency, including two siblings, have been identified so far, showing accumulation of the C27-bile acid intermediates. Here, we performed biochemical studies in material from a novel ACOX2-deficient patient with increased levels of C27-bile acids in plasma, a complete loss of ACOX2 protein expression on immunoblot, but normal pristanic acid oxidation activity in fibroblasts. Since pristanoyl-CoA is presumed to be handled by ACOX2 specifically, these findings prompted us to re-investigate the expression of the human peroxisomal acyl-CoA oxidases. We report for the first time expression of ACOX3 in normal human tissues at the mRNA and protein level. Substrate specificity studies were done for ACOX1, 2 and 3 which revealed that ACOX1 is responsible for the oxidation of straight-chain fatty acids with different chain lengths, ACOX2 is the only human acyl-CoA oxidase involved in bile acid biosynthesis, and both ACOX2 and ACOX3 are involved in the degradation of the branched-chain fatty acids. Our studies provide new insights both into ACOX2 deficiency and into the role of the different acyl-CoA oxidases in peroxisomal metabolism.
Collapse
Affiliation(s)
- Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands.
| | - Simone Denis
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Carlo W T van Roermund
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Mary Anne Preece
- Department of Newborn Screening and Biochemical Genetics, Birmingham Children's Hospital NHS Foundation Trust, Birmingham B4 6NH, United Kingdom
| | - Janet Koster
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Merel S Ebberink
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Abstract
Due to a peculiar age-dependent increased susceptibility, neonatal cholestasis affects the liver of approximately 1 in every 2500 term infants. A high index of suspicion is the key to an early diagnosis, and to implement timely, often life-saving treatments. Even when specific treatment is not available or curative, prompt medical management and optimization of nutrition are of paramount importance to survival and avoidance of complications. Areas covered: The present article will prominently focus on a series of newer diagnostic and therapeutic options of cholestasis in neonates and infants blended with consolidated established paradigms. The overview of strategies for the management reported here is based on a systematic literature search published in English using accessible databases (PubMed, MEDLINE) with the keywords biliary atresia, choleretics and neonatal cholestasis. References lists from retrieved articles were also reviewed. Expert commentary: A large number of uncommon and rare hepatobiliary disorders may present with cholestasis during the neonatal and infantile period. Potentially life-saving disease-specific pharmacological and surgical therapeutic approaches are currently available. Advances in hepatobiliary transport mechanisms have started clarifying fundamental aspects of inherited and acquired cholestasis, laying the foundation for the development of possibly more effective specific therapies.
Collapse
Affiliation(s)
- Andrea Catzola
- a Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Pediatrics Section , University of Salerno , Salerno , Italy
| | - Pietro Vajro
- a Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Pediatrics Section , University of Salerno , Salerno , Italy
| |
Collapse
|
37
|
|
38
|
Vaz FM, Ferdinandusse S. Bile acid analysis in human disorders of bile acid biosynthesis. Mol Aspects Med 2017; 56:10-24. [PMID: 28322867 DOI: 10.1016/j.mam.2017.03.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/11/2017] [Accepted: 03/16/2017] [Indexed: 01/17/2023]
Abstract
Bile acids facilitate the absorption of lipids in the gut, but are also needed to maintain cholesterol homeostasis, induce bile flow, excrete toxic substances and regulate energy metabolism by acting as signaling molecules. Bile acid biosynthesis is a complex process distributed across many cellular organelles and requires at least 17 enzymes in addition to different metabolite transport proteins to synthesize the two primary bile acids, cholic acid and chenodeoxycholic acid. Disorders of bile acid synthesis can present from the neonatal period to adulthood and have very diverse clinical symptoms ranging from cholestatic liver disease to neuropsychiatric symptoms and spastic paraplegias. This review describes the different bile acid synthesis pathways followed by a summary of the current knowledge on hereditary disorders of human bile acid biosynthesis with a special focus on diagnostic bile acid profiling using mass spectrometry.
Collapse
Affiliation(s)
- Frédéric M Vaz
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands.
| | - Sacha Ferdinandusse
- Department of Clinical Chemistry and Pediatrics, Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|