Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Cell Biol Toxicol ; 40(1): 62, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093497

ABSTRACT

BACKGROUND: Increased activity of the transcription factor FOXC1 leads to elevated transcription of target genes, ultimately facilitating the progression of various cancer types. However, there are currently no literature reports on the role of FOXC1 in renal cell carcinoma. METHODS: By using RT-qPCR, immunohistochemistry and Western blotting, FOXC1 mRNA and protein expression was evaluated. Gain of function experiments were utilized to assess the proliferation and metastasis ability of cells. A nude mouse model was created for transplanting tumors and establishing a lung metastasis model to observe cell proliferation and spread in a living organism. Various techniques including biological analysis, CHIP assay, luciferase assay, RT-qRCR and Western blotting experiments were utilized to investigate how FOXC1 contributes to the transcription of ABHD5 on a molecular level. FOXC1 was assessed by Western blot for its impact on AMPK/mTOR signaling pathway. RESULTS: FOXC1 is down-regulated in RCC, causing unfavorable prognosis of patients with RCC. Further experiments showed that forced FOXC1 expression significantly restrains RCC cell growth and cell metastasis. Mechanically, FOXC1 promotes the transcription of ABHD5 to activate AMPK signal pathway to inhibit mTOR signal pathway. Finally, knockdown of ABHD5 recovered the inhibitory role of FOXC1 overexpression induced cell growth and metastasis suppression. CONCLUSION: In general, our study demonstrates that FOXC1 exerts its tumor suppressor role by promoting ABHD5 transcription to regulating AMPK/mTOR signal pathway. FOXC1 could serve as both a diagnostic indicator and potential treatment focus for RCC.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , AMP-Activated Protein Kinases , Carcinoma, Renal Cell , Cell Proliferation , Forkhead Transcription Factors , Kidney Neoplasms , Mice, Nude , Signal Transduction , TOR Serine-Threonine Kinases , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Humans , Animals , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Cell Line, Tumor , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Cell Proliferation/genetics , Mice , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Gene Expression Regulation, Neoplastic , Disease Progression , Male , Female , Mice, Inbred BALB C
2.
Am J Physiol Endocrinol Metab ; 327(1): E69-E80, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717361

ABSTRACT

Acylglycerophosphate acyltransferases (AGPATs) catalyze the de novo formation of phosphatidic acid to synthesize glycerophospholipids and triglycerides. AGPATs demonstrate unique physiological roles despite a similar biochemical function. AGPAT3 is highly expressed in the testis, kidney, and liver, with intermediate expression in adipose tissue. Loss of AGPAT3 is associated with reproductive abnormalities and visual dysfunction. However, the role of AGPAT3 in adipose tissue and whole body metabolism has not been investigated. We found that male Agpat3 knockout (KO) mice exhibited reduced body weights with decreased white and brown adipose tissue mass. Such changes were less pronounced in the female Agpat3-KO mice. Agpat3-KO mice have reduced plasma insulin growth factor 1 (IGF1) and insulin levels and diminished circulating lipid metabolites. They manifested intact glucose homeostasis and insulin sensitivity despite a lean phenotype. Agpat3-KO mice maintained an energy balance with normal food intake, energy expenditure, and physical activity, except for increased water intake. Their adaptive thermogenesis was also normal despite reduced brown adipose mass and triglyceride content. Mechanistically, Agpat3 was elevated during mouse and human adipogenesis and enriched in adipocytes. Agpat3-knockdown 3T3-L1 cells and Agpat3-deficient mouse embryonic fibroblasts (MEFs) have impaired adipogenesis in vitro. Interestingly, pioglitazone treatment rescued the adipogenic deficiency in Agpat3-deficient cells. We conclude that AGPAT3 regulates adipogenesis and adipose development. It is possible that adipogenic impairment in Agpat3-deficient cells potentially leads to reduced adipose mass. Findings from this work support the unique role of AGPAT3 in adipose tissue.NEW & NOTEWORTHY AGPAT3 deficiency results in male-specific growth retardation. It reduces adipose tissue mass but does not significantly impact glucose homeostasis or energy balance, except for influencing water intake in mice. Like AGPAT2, AGPAT3 is upregulated during adipogenesis, potentially by peroxisome proliferator-activated receptor gamma (PPARγ). Loss of AGPAT3 impairs adipocyte differentiation, which could be rescued by pioglitazone. Overall, AGPAT3 plays a significant role in regulating adipose tissue mass, partially involving its influence on adipocyte differentiation.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Adipocytes , Mice, Knockout , Animals , Female , Male , Mice , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Cell Differentiation , Energy Metabolism/genetics , Insulin Resistance/genetics , Mice, Inbred C57BL , Phenotype , Thermogenesis/genetics , Thinness/metabolism , Thinness/genetics
3.
Mol Microbiol ; 121(6): 1164-1181, 2024 06.
Article in English | MEDLINE | ID: mdl-38676355

ABSTRACT

Latent tuberculosis, caused by dormant Mycobacterium tuberculosis (Mtb), poses a threat to global health through the incubation of undiagnosed infections within the community. Dormant Mtb, which is phenotypically tolerant to antibiotics, accumulates triacylglycerol (TAG) utilizing fatty acids obtained from macrophage lipid droplets. TAG is vital to mycobacteria, serving as a cell envelope component and energy reservoir during latency. TAG synthesis occurs by sequential acylation of glycerol-3-phosphate, wherein the second acylation step is catalyzed by acylglycerol-3-phosphate acyltransferase (AGPAT), resulting in the production of phosphatidic acid (PA), a precursor for the synthesis of TAG and various phospholipids. Here, we have characterized a putative acyltransferase of Mtb encoded by Rv3816c. We found that Rv3816c has all four characteristic motifs of AGPAT, exists as a membrane-bound enzyme, and functions as 1-acylglycerol-3-phosphate acyltransferase. The enzyme could transfer the acyl group to acylglycerol-3-phosphate (LPA) from monounsaturated fatty acyl-coenzyme A of chain length 16 or 18 to produce PA. Complementation of Escherichia coli PlsC mutant in vivo by Rv3816c confirmed that it functions as AGPAT. Its active site mutants, H43A and D48A, were incapable of transferring the acyl group to LPA in vitro and were not able to rescue the growth defect of E. coli PlsC mutant in vivo. Identifying Rv3816c as AGPAT and comparing its properties with other AGPAT homologs is not only a step toward understanding the TAG biosynthesis in mycobacteria but has the potential to explore it as a drug target.


Subject(s)
Mycobacterium tuberculosis , Triglycerides , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Triglycerides/biosynthesis , Triglycerides/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Glycerol-3-Phosphate O-Acyltransferase/genetics , Acyltransferases/metabolism , Acyltransferases/genetics , Acylation , Fatty Acids/metabolism , Fatty Acids/biosynthesis , Phosphatidic Acids/metabolism , Phosphatidic Acids/biosynthesis , Acyl Coenzyme A/metabolism
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167007, 2024 03.
Article in English | MEDLINE | ID: mdl-38185063

ABSTRACT

The development of nonalcoholic fatty liver disease (NAFLD) may worsen due to chronic stress or prolonged use of glucocorticoids. Glycerol-3-phosphate acyltransferase 3 (GPAT3), has a function in obesity and serves as a key rate-limiting enzyme that regulates triglyceride synthesis. However, the precise impact of GPAT3 on corticosterone (CORT)-induced NAFLD and its underlying molecular mechanism remain unclear. For our in vivo experiments, we utilized male and female mice that were GPAT3-/- and wild type (WT) and treated them with CORT for a duration of 4 weeks. In our in vitro experiments, we transfected AML12 cells with GPAT3 siRNA and subsequently treated them with CORT. Under CORT-treated conditions, the absence of GPAT3 greatly improved obesity and hepatic steatosis while enhancing the expression of genes involved in fatty acid oxidation, as evidenced by our findings. In addition, the deletion of GPAT3 significantly inhibited the production of reactive oxygen species (ROS), increased the expression of antioxidant genes, and recovered the mitochondrial membrane potential in AML12 cells treated with CORT. In terms of mechanism, the absence of GPAT3 encouraged the activation of the glycogen synthase kinase 3ß (GSK3ß)/nuclear factor-erythroid 2 related factor 2 (Nrf2) pathway, which served as a defense mechanism against liver fat accumulation and oxidative stress. Furthermore, GPAT3 expression was directly controlled at the transcriptional level by the glucocorticoid receptor (GR). Collectively, our findings suggest that GPAT3 deletion significantly alleviated hepatic steatosis and oxidative stress through promoting GSK3ß/Nrf2 signaling pathways.


Subject(s)
Non-alcoholic Fatty Liver Disease , Male , Female , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Corticosterone/pharmacology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Oxidative Stress , Obesity/drug therapy , Obesity/genetics , Acyltransferases/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism
5.
Arch Med Res ; 55(1): 102925, 2024 01.
Article in English | MEDLINE | ID: mdl-38042031

ABSTRACT

BACKGROUND AND AIM: Gestational diabetes mellitus (GDM) is one of the most common metabolic disorders in pregnancy, and a novel association of maternal lipid profile has been suggested to play an important role. However, the molecular mechanism is not clear. METHODS: Bio-analyzed combined with placental metabonomics and single-cell RNA-sequencing (scRNA-seq) successfully identified a potentially important molecule: α-ß hydrolase domain-containing protein 5 (ABHD5). The syncytiotrophoblast (SCT) cell model was adopted as a fusion of BeWo cells in response to forskolin. On this basis, the high glucose-stimulated cell experiment was carried out. 15 women with GDM and 15 normal pregnant women were recruited for validation experiments. RESULTS: ABHD5 was mainly expressed in the trophoblast cells, especially in SCT cells, and significantly decreased in the GDM placenta. After stimulation by high glucose, the expression of ABHD5 was downregulated in a time-dependent manner in BeWo cells treated with forskolin. At the same time, lipid droplets (LDs) were increased in the SCT. LD storage was also increased in the SCT with siABHD5, while it was significantly reduced in SCT cells with high ABHD5 expression. However, this effect could be attenuated by downregulated carnitine palmitoyltransferase 1B (CPT1B). CONCLUSIONS: ABHD5-CPT1B is confirmed as an important regulator of placental lipid metabolism.


Subject(s)
Diabetes, Gestational , Placenta , Female , Humans , Pregnancy , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Colforsin/pharmacology , Colforsin/metabolism , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Glucose/metabolism , Lipid Metabolism , Placenta/metabolism
6.
J Clin Lipidol ; 18(1): e125-e128, 2024.
Article in English | MEDLINE | ID: mdl-37968200

ABSTRACT

Chanarin-Dorfman Syndrome (CDS) is a rare lipid storage disease with ichthyosis, hepatomegaly, myopathy, neuropathy, deafness, and ocular findings. Here, we aim to present an elderly CDS case and highlight the new endocrinological findings. A 66-year-old male patient with cirrhosis was hospitalized for liver transplantation. We suspected Chanarin-Dorfman Syndrome with ichthyosis, fatty liver, and syndromic facial features with bilateral ectropion, deafness, and malocclusion. We showed the lipid droplets in neutrophils called patognomonic Jordans' anomaly. Homozygous c.47+1 G>A mutation in the ABHD5 (NM_016006.6) gene were detected by clinical exome sequencing. Out of <160 CDS cases in the literature, this is the second eldest CDS patient and first with adrenal insufficiency, parathyroid lipoadenoma and atrophic pancreas. Clinicians should be aware of CDS as a rare cause of fatty liver. We recommend a blood smear and genetic analyses in patients with severe ichtiosis, ectropion, deafness and multiple endocrinolgic disorders.


Subject(s)
Deafness , Ectropion , Fatty Liver , Ichthyosiform Erythroderma, Congenital , Ichthyosis , Lipid Metabolism, Inborn Errors , Liver Transplantation , Muscular Diseases , Male , Humans , Aged , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Fatty Liver/diagnosis , Fatty Liver/genetics , Lipids , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics
7.
Am J Med Genet A ; 194(4): e63481, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37984424

ABSTRACT

Chanarin-Dorfman syndrome is an autosomal recessively inherited disorder characterized by ichthyosis, sensorineural hearing loss, and hepatic dysfunction. We report on a 60-year-old female of Venezuelan descent who presented with congenital ichthyosis, progressive sensorineural hearing loss, and liver cirrhosis. We identify a heterozygous copy number deletion involving exon 1 and another heterozygous deletion involving exon 3 of the ABHD5 gene. Exon 2 is preserved. Both deletions were confirmed with RT-PCR. RNAseq from peripheral blood shows a reduction of ABHD5 expression overall and an absence of exon 3 expression, confirming the deleterious effects of the identified deletions. We present exonic deletions as a potentially common type of ABHD5 variation.


Subject(s)
Hearing Loss, Sensorineural , Ichthyosiform Erythroderma, Congenital , Ichthyosis , Lipid Metabolism, Inborn Errors , Muscular Diseases , Female , Humans , Middle Aged , Ichthyosiform Erythroderma, Congenital/complications , Ichthyosiform Erythroderma, Congenital/diagnosis , Ichthyosiform Erythroderma, Congenital/genetics , Lipid Metabolism, Inborn Errors/genetics , Muscular Diseases/genetics , Ichthyosis/complications , Ichthyosis/diagnosis , Ichthyosis/genetics , Liver Cirrhosis , Hearing Loss, Sensorineural/complications , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics
8.
Proc Natl Acad Sci U S A ; 120(52): e2312666120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38127985

ABSTRACT

AGPAT2 (1-acyl-sn-glycerol-3-phosphate-acyltransferase-2) converts lysophosphatidic acid (LPA) into phosphatidic acid (PA), and mutations of the AGPAT2 gene cause the most common form of congenital generalized lipodystrophy which leads to steatohepatitis. The underlying mechanism by which AGPAT2 deficiency leads to lipodystrophy and steatohepatitis has not been elucidated. We addressed this question using an antisense oligonucleotide (ASO) to knockdown expression of Agpat2 in the liver and white adipose tissue (WAT) of adult male Sprague-Dawley rats. Agpat2 ASO treatment induced lipodystrophy and inflammation in WAT and the liver, which was associated with increased LPA content in both tissues, whereas PA content was unchanged. We found that a controlled-release mitochondrial protonophore (CRMP) prevented LPA accumulation and inflammation in WAT whereas an ASO against glycerol-3-phosphate acyltransferase, mitochondrial (Gpam) prevented LPA content and inflammation in the liver in Agpat2 ASO-treated rats. In addition, we show that overnutrition, due to high sucrose feeding, resulted in increased hepatic LPA content and increased activated macrophage content which were both abrogated with Gpam ASO treatment. Taken together, these data identify LPA as a key mediator of liver and WAT inflammation and lipodystrophy due to AGPAT2 deficiency as well as liver inflammation due to overnutrition and identify LPA as a potential therapeutic target to ameliorate these conditions.


Subject(s)
Fatty Liver , Lipodystrophy , Overnutrition , Male , Rats , Animals , Acyltransferases/metabolism , Glycerol , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Rats, Sprague-Dawley , Lipodystrophy/genetics , Adipose Tissue, White/metabolism , Phosphatidic Acids , Inflammation , Phosphates
9.
Genes (Basel) ; 14(11)2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38003015

ABSTRACT

During triacylglycerol synthesis, the acylglycerol-3-phosphate acyltransferase (AGPAT) family catalyzes the conversion of lysophosphatidic acid to phosphatidic acid and the acylation of sn-2 fatty acids. However, the catalytic activity of different AGPAT members is different. Therefore, this study aimed to investigate the mechanism through which different AGPATs affect the efficiency of TAG synthesis and fatty acid composition. The conservation of amino acid sequences and protein domains of the AGPAT family was analyzed, and the functions of AGPAT1, AGPAT3, and AGPAT4 genes in buffalo mammary epithelial cells (BMECs) were studied using RNA interference and gene overexpression. Prediction of the protein tertiary structure of the AGPAT family demonstrated that four conservative motifs (motif1, motif2, motif3, and motif6) formed a hydrophobic pocket in AGPAT proteins, except AGPAT6. According to cytological studies, AGPAT1, AGPAT3, and AGPAT4 were found to promote the synthesis and fatty acid compositions of triacylglycerol, especially UFA compositions of triacylglycerol, by regulating ACSL1, FASN, GPAM, DGAT2, and PPARG gene expression. This study provides new insights into the role of different AGPAT gene family members involved in TAG synthesis, and a reference for improving the fatty acid composition of milk.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Buffaloes , Animals , Buffaloes/genetics , Buffaloes/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Milk/metabolism , Fatty Acids/genetics , Triglycerides
10.
Metabolism ; 148: 155693, 2023 11.
Article in English | MEDLINE | ID: mdl-37741434

ABSTRACT

BACKGROUND & AIMS: Autophagy-related 14 (ATG14) is a key regulator of autophagy. ATG14 is also localized to lipid droplet; however, the function of ATG14 on lipid droplet remains unclear. In this study, we aimed to elucidate the role of ATG14 in lipid droplet homeostasis. METHODS: ATG14 loss-of-function and gain-of-function in lipid droplet metabolism were analyzed by fluorescence imaging in ATG14 knockdown or overexpression hepatocytes. Specific domains involved in the ATG14 targeting to lipid droplets were analyzed by deletion or site-specific mutagenesis. ATG14-interacting proteins were analyzed by co-immunoprecipitation. The effect of ATG14 on lipolysis was analyzed in human hepatocytes and mouse livers that were deficient in ATG14, comparative gene identification-58 (CGI-58), or both. RESULTS: Our data show that ATG14 is enriched on lipid droplets in hepatocytes. Mutagenesis analysis reveals that the Barkor/ATG14 autophagosome targeting sequence (BATS) domain of ATG14 is responsible for the ATG14 localization to lipid droplets. Co-immunoprecipitation analysis illustrates that ATG14 interacts with adipose triglyceride lipase (ATGL) and CGI-58. Moreover, ATG14 also enhances the interaction between ATGL and CGI-58. In vitro lipolysis analysis demonstrates that ATG14 deficiency remarkably decreases triglyceride hydrolysis. CONCLUSIONS: Our data suggest that ATG14 can directly enhance lipid droplet breakdown through interactions with ATGL and CGI-58.


Subject(s)
Lipase , Lipid Droplets , Mice , Animals , Humans , Lipase/metabolism , Lipid Droplets/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Lipolysis , Lipid Metabolism/genetics , Liver/metabolism , Homeostasis , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism
11.
Front Immunol ; 14: 1026669, 2023.
Article in English | MEDLINE | ID: mdl-36845084

ABSTRACT

Background: Liver cancer is the sixth most commonly diagnosed cancer and the third leading cause of cancer-related death worldwide. Hepatocellular carcinoma accounts for an estimated 90% of all liver cancers. Many enzymes of the GPAT/AGPAT family are required for the synthesis of triacylglycerol. Expression of AGPAT isoenzymes has been reported to be associated with an increased risk of tumorigenesis or development of aggressive phenotypes in a variety of cancers. However, whether members of the GPAT/AGPAT gene family also influence the pathophysiology of HCC is unknown. Methods: Hepatocellular carcinoma datasets were obtained from the TCGA and ICGC databases. Predictive models related to the GPAT/AGPAT gene family were constructed based on LASSO-Cox regression using the ICGC-LIRI dataset as an external validation cohort. Seven immune cell infiltration algorithms were used to analyze immune cell infiltration patterns in different risk groups. IHC, CCK-8, Transwell assay, and Western blotting were used for in vitro validation. Results: Compared with low-risk patients, high-risk patients had shorter survival and higher risk scores. Multivariate Cox regression analysis showed that risk score was a significant independent predictor of overall survival (OS) after adjustment for confounding clinical factors (p < 0.001). The established nomogram combined risk score and TNM staging to accurately predict survival at 1, 3, and 5 years in patients with HCC with AUC values of 0.807, 0.806, and 0.795, respectively. This risk score improved the reliability of the nomogram and guided clinical decision-making. In addition, we comprehensively analyzed immune cell infiltration (using seven algorithms), response to immune checkpoint blockade, clinical relevance, survival, mutations, mRNA expression-based stemness index, signaling pathways, and interacting proteins related to the three core genes of the prognostic model (AGPAT5, LCLAT1, and LPCAT1). We also performed preliminary validation of the differential expression, oncological phenotype, and potential downstream pathways of the three core genes by IHC, CCK-8, Transwell assay, and Western blotting. Conclusion: These results improve our understanding of the function of GPAT/AGPAT gene family members and provide a reference for prognostic biomarker research and individualized treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Prognosis , Reproducibility of Results , Sincalide , Tumor Microenvironment/genetics , Glycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics
12.
Pediatr Dermatol ; 40(5): 879-881, 2023.
Article in English | MEDLINE | ID: mdl-36709747

ABSTRACT

Chanarin-Dorfman syndrome (CDS) is a rare, autosomal recessive disorder of impaired triacylglycerol catabolism leading to cytoplasmic deposition of triglycerides in various cell types. We describe the case of an 8-month-old boy with cataracts, strabismus, motor delays, and an ichthyosiform rash since birth. Genetic testing revealed a pathogenic variant of the ABHD5 gene, suggestive of CDS, and further workup demonstrated hepatic steatosis and myopathy. His ichthyosis improved with initiation of a diet low in very long-chain fatty acids and medium-chain fatty acid supplementation.


Subject(s)
Cataract , Ichthyosiform Erythroderma, Congenital , Ichthyosis, Lamellar , Ichthyosis , Lipid Metabolism, Inborn Errors , Muscular Diseases , Male , Humans , Infant , Ichthyosiform Erythroderma, Congenital/diagnosis , Ichthyosiform Erythroderma, Congenital/genetics , Ichthyosis, Lamellar/diagnosis , Ichthyosis, Lamellar/genetics , Ichthyosis/diagnosis , Ichthyosis/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/pathology , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Cataract/diagnosis , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics
13.
Cell Biochem Funct ; 41(1): 128-137, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36515301

ABSTRACT

Dysfunction of the adipose tissue metabolism is considered as a significant hallmark of aging. It has been proposed that α-ß hydrolase domain containing 5 (ABHD5) plays a critical role in the control of lipolysis. However, the role of ABHD5 in the control of lipolysis during aging or exercise is unknown. Here we combined the experimental mouse model with transcriptomic analyzes by using murine and human databases to explore the role of ABHD5 in the adipose tissue during aging and in response to exercise. Transcriptomic data revealed a downregulation of Abhd5 messenger RNA levels in the subcutaneous white adipose tissue (scWAT) over time in individuals from 20 to 69 years old. Aged mice displayed dramatic reduction of ABHD5 protein content and lipolytic-related proteins in the scWAT. Interestingly, 4 weeks of high-intensity interval training increased ABHD5 protein level and restored the lipolytic pathway in the scWAT of aged mice. Altogether, our findings demonstrated that aging affects ABHD5 content in the adipose tissue of mice and humans. Conversely, exercise increases ABHD5 activity, recovering the lipolytic activity in aged mice.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Adipose Tissue , Aging , Exercise , Lipolysis , Adult , Aged , Animals , Humans , Mice , Middle Aged , Young Adult , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Adipose Tissue/enzymology , Aging/metabolism , Hydrolases/genetics , Hydrolases/metabolism
14.
Cancer Biother Radiopharm ; 38(5): 336-346, 2023 Jun.
Article in English | MEDLINE | ID: mdl-32822232

ABSTRACT

Background: Lung adenocarcinoma (LAC) is a common malignancy worldwide. Emerging findings indicated that circular RNAs possess complex capacities of gene modulation in tumorigenesis and metastasis. Nevertheless, the role of circular RNA in LAC is still largely unknown. Materials and Methods: The level of circular RNA cMras (circ_cMras), alpha-beta hydrolase domain 5 (ABHD5), and adipose triglyceride lipase (ATGL) was determined by quantitative real-time polymerase chain reaction assay. Protein levels of ABHD5, ATGL, p53, p65, and phospho-p65 (p-p65) were examined by Western blot. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was used to detect cell proliferation in vitro. Cell apoptosis was estimated using flow cytometry. Transwell assay was used to measure cell migration and invasion in A549 and HCC827 cells. Finally, the role of circ_cMras was explored using xenograft tumor model. Results: Low levels of circ_cMras, ABHD5, and ATGL were observed in LAC tissues and cells. Upregulation of circ_cMras could hamper tumor aggression in vitro and in vivo, exhibiting as the inhibition of cell proliferation, migration, invasion, and promotion of cell apoptosis, as well as the inhibition on tumor growth in vivo. Moreover, ABHD5 deletion could overturn the effects of circ_cMras overexpression on cell behaviors in LAC cells. Furthermore, the inhibiting effects of ABHD5 on cell aggression were reversed by ATGL deficiency in vitro. Mechanically, circ_cMras/ABHD5/ATGL axis exerted its role through NF-κB signaling pathway in LAC cells. Conclusion: Circ_cMras exerted its function through ABHD5/ATGL axis using NF-κB signaling pathway in LAC, which might provide a novel insight for the diagnosis and prognosis of LAC.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , MicroRNAs , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , NF-kappa B/metabolism , Hydrolases/metabolism , Cell Line, Tumor , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Signal Transduction , Lung Neoplasms/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Apoptosis/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism
15.
J Mol Cell Biol ; 14(8)2022 12 26.
Article in English | MEDLINE | ID: mdl-36107452

ABSTRACT

Comparative gene identification-58 (CGI-58), also known as α/ß hydrolase domain containing 5, is the co-activator of adipose triglyceride lipase that hydrolyzes triglycerides stored in the cytosolic lipid droplets. Mutations in CGI-58 gene cause Chanarin-Dorfman syndrome (CDS), an autosomal recessive neutral lipid storage disease with ichthyosis. The liver pathology of CDS manifests as steatosis and steatohepatitis, which currently has no effective treatments. Perilipin-3 (Plin3) is a member of the Perilipin-ADRP-TIP47 protein family that is essential for lipid droplet biogenesis. The objective of this study was to test a hypothesis that deletion of a major lipid droplet protein alleviates fatty liver pathogenesis caused by CGI-58 deficiency in hepatocytes. Adult CGI-58-floxed mice were injected with adeno-associated vectors simultaneously expressing the Cre recombinase and microRNA against Plin3 under the control of a hepatocyte-specific promoter, followed by high-fat diet feeding for 6 weeks. Liver and blood samples were then collected from these animals for histological and biochemical analysis. Plin3 knockdown in hepatocytes prevented steatosis, steatohepatitis, and necroptosis caused by hepatocyte CGI-58 deficiency. Our work is the first to show that inhibiting Plin3 in hepatocytes is sufficient to mitigate hepatocyte CGI-58 deficiency-induced hepatic steatosis and steatohepatitis in mice.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Fatty Liver , Mice , Animals , Perilipin-3 , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Hepatocytes/metabolism , Triglycerides/metabolism
16.
J Dermatol Sci ; 107(2): 89-94, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35970721

ABSTRACT

BACKGROUND: PNPLA1 is a causative gene of autosomal recessive congenital ichthyosis. The transacylase PNPLA1 produces ω-O-acylceramides (acylceramides), lipids essential for the skin barrier function, by catalyzing the transfer of a linoleic acid from triglycerides to ω-hydroxyceramides. OBJECTIVE: We aimed to validate the involvement of PNPLA1 mutations found in ichthyosis patients in the pathogenesis and elucidate the correlation between the effects of these mutations on acylceramide-producing activity and ichthyosis pathology. METHODS: Acylceramide-producing activity of PNPLA1 mutants was investigated using a cell-based assay system, in which wild-type PNPLA1 or each PNPLA1 mutant was co-overexpressed with the enzymes involved in acylceramide synthesis. The effect of each mutation on the ABHD5-dependent lipid droplet localization of PNPLA1 was examined through indirect immunofluorescence microscopy. RESULTS: Of 16 PNPLA1 missense mutations, 15 mutations, except the C216R mutation, resulted in a complete loss of acylceramide-producing activity, while the C216R mutation weakly affected this activity. Intracellular localization of mutants with no activity varied among mutants. Two mutants (S19L and D172N) localized in lipid droplets, and eight mutants (S53L, S53W, A59V, T125N, D129E, R166C, P234S, and P235L) partially localized there. Five mutants (A34P, A34T, S53P, K141E, and P163L) localized throughout the cytosol. CONCLUSION: The PNPLA1 missense mutations examined in this study are responsible for ichthyosis pathology. The weak effect of C216R mutation on acylceramide-producing activity correlates with the mild symptoms of the ichthyosis patient. Sixteen PNPLA1 mutants were classified into four groups based on their acylceramide-producing activity and localization.


Subject(s)
Ichthyosis, Lamellar , Ichthyosis , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Acyltransferases , Ceramides , Humans , Ichthyosis/genetics , Ichthyosis, Lamellar/genetics , Linoleic Acid , Lipase/genetics , Mutation , Skin , Triglycerides
17.
Sci Rep ; 12(1): 2565, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35173175

ABSTRACT

Alpha/beta hydrolase domain-containing protein 5 (ABHD5) is a highly conserved protein that regulates various lipid metabolic pathways via interactions with members of the perilipin (PLIN) and Patatin-like phospholipase domain-containing protein (PNPLA) protein families. Loss of function mutations in ABHD5 result in Chanarin-Dorfman Syndrome (CDS), characterized by ectopic lipid accumulation in numerous cell types and severe ichthyosis. Recent data demonstrates that ABHD5 is the target of synthetic and endogenous ligands that might be therapeutic beneficial for treating metabolic diseases and cancers. However, the structural basis of ABHD5 functional activities, such as protein-protein interactions and ligand binding is presently unknown. To address this gap, we constructed theoretical structural models of ABHD5 by comparative modeling and topological shape analysis to assess the spatial patterns of ABHD5 conformations computed in protein dynamics. We identified functionally important residues on ABHD5 surface for lipolysis activation by PNPLA2, lipid droplet targeting and PLIN-binding. We validated the computational model by examining the effects of mutating key residues in ABHD5 on an array of functional assays. Our integrated computational and experimental findings provide new insights into the structural basis of the diverse functions of ABHD5 as well as pathological mutations that result in CDS.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/chemistry , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Computational Biology/methods , Lipase/metabolism , Lipid Droplets/metabolism , Mutation , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Humans , Ligands , Lipid Droplets/chemistry , Protein Conformation
18.
G3 (Bethesda) ; 12(2)2022 02 04.
Article in English | MEDLINE | ID: mdl-34791225

ABSTRACT

Ichthyoses are hereditary skin disorders characterized by the formation of scales and defects in the outermost layer of the epidermis. In dogs, at least six different breed-specific ichthyoses including a relatively common PNPLA1-related autosomal recessive ichthyosis in Golden Retrievers are known. In this study, we investigated 14 Golden Retrievers with scales that were not homozygous for the mutant PNPLA1 allele suggesting a genetically distinct new form of ichthyosis. Histopathological examinations showed lamellar, orthokeratotic hyperkeratosis, and mildly hyperplastic epidermis that led to the diagnosis of a nonepidermolytic ichthyosis. Combined linkage and homozygosity mapping in 14 cases and 30 nonaffected family members delimited a critical interval of ∼12.7 Mb on chromosome 23. Whole-genome sequencing of an affected dog revealed a single protein-changing variant within this region that was not present in 795 control genomes. The identified variant is a 14 bp deletion in the ABHD5 gene (c.1006_1019del), leading to a frameshift and altering the last 14 codons p.(Asp336Serfs*6). The genotypes at this variant showed perfect cosegregation with the ichthyosis phenotype in a large family comprising 14 cases and 72 controls. ABHD5 encodes an acyltransferase required for lipid metabolism. In humans, variants in ABHD5 cause Chanarin-Dorfman syndrome, a neutral lipid storage disease with ichthyosis. Our data in dogs together with the knowledge on the effects of ABHD5 variants in humans strongly suggest ABHD5:c.1006_1019del as candidate causative genetic variant for a new canine form of ichthyosis, which we propose to designate as Golden Retriever ichthyosis type 2 (ICH2).


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase , Ichthyosiform Erythroderma, Congenital , Ichthyosis, Lamellar , Ichthyosis , Lipid Metabolism, Inborn Errors , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Animals , Dogs , Frameshift Mutation , Gene Deletion , Ichthyosiform Erythroderma, Congenital/genetics , Ichthyosiform Erythroderma, Congenital/pathology , Ichthyosis/genetics , Ichthyosis/pathology , Ichthyosis/veterinary , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/veterinary , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Plant Breeding
19.
Nat Commun ; 12(1): 6711, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795238

ABSTRACT

Cancer stemness represents a major source of development and progression of colorectal cancer (CRC). c-Met critically contributes to CRC stemness, but how c-Met is activated in CRC remains elusive. We previously identified the lipolytic factor ABHD5 as an important tumour suppressor gene in CRC. Here, we show that loss of ABHD5 promotes c-Met activation to sustain CRC stemness in a non-canonical manner. Mechanistically, we demonstrate that ABHD5 interacts in the cytoplasm with the core subunit of the SET1A methyltransferase complex, DPY30, thereby inhibiting the nuclear translocation of DPY30 and activity of SET1A. In the absence of ABHD5, DPY30 translocates to the nucleus and supports SET1A-mediated methylation of YAP and histone H3, which sequesters YAP in the nucleus and increases chromatin accessibility to synergistically promote YAP-induced transcription of c-Met, thus promoting the stemness of CRC cells. This study reveals a novel role of ABHD5 in regulating histone/non-histone methylation and CRC stemness.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-met/genetics , YAP-Signaling Proteins/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , HCT116 Cells , Humans , Male , Methylation , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Proto-Oncogene Proteins c-met/metabolism , Pyrazines/pharmacology , Triazines/pharmacology , Xenograft Model Antitumor Assays/methods , YAP-Signaling Proteins/metabolism
20.
Genes (Basel) ; 12(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440338

ABSTRACT

Autosomal recessive Chanarin-Dorfman syndrome (CDS, MIM #275630) is defined as a neutral lipid storage disease with ichthyosis (NLSDI) due to an accumulation of lipid droplets in a variety of different tissues including liver and muscle cells, leucocytes, fibroblasts and nerve cells It is caused by biallelic mutations in the abhydrolase domain containing 5 gene (ABHD5, MIM *604780) which is localized on the short arm of chromosome 3. Here we report an 18 month-old girl in whom we have identified the homozygous ABHD5 mutation c.700C > T, p.(Arg234*). Since none of the parents carried this point mutation, parentage was confirmed by microsatellite marker analysis. Suspected uniparental disomy (UPD) was confirmed by microsatellite genotyping over the entire chromosome 3 and indicated a maternal origin. UPD is an extremely rare event that is not necessarily pathogenic, but may cause disease if the affected chromosome contains genes that are imprinted. Here we report the first case of Chanarin-Dorfman syndrome due to a de novo ABHD5 mutation in the maternal germ cell, combined with a maternal uniparental isodisomy of chromosome 3. This case demonstrates that genetic analysis of the patient and both parents is crucial to provide correct genetic counseling.


Subject(s)
1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Chromosomes, Human, Pair 3 , Genes, Recessive , Ichthyosiform Erythroderma, Congenital/genetics , Lipid Metabolism, Inborn Errors/genetics , Muscular Diseases/genetics , Mutation , Female , Homozygote , Humans , Infant
SELECTION OF CITATIONS
SEARCH DETAIL