Original Article Rosiglitazone Inhibits Acyl-Coa Synthetase Activity and Fatty Acid

Total Page:16

File Type:pdf, Size:1020Kb

Original Article Rosiglitazone Inhibits Acyl-Coa Synthetase Activity and Fatty Acid Original Article Rosiglitazone Inhibits Acyl-CoA Synthetase Activity and Fatty Acid Partitioning to Diacylglycerol and Triacylglycerol via a Peroxisome Proliferator–Activated Receptor-␥–Independent Mechanism in Human Arterial Smooth Muscle Cells and Macrophages Bardia Askari,1 Jenny E. Kanter,1 Ashley M. Sherrid,1 Deidre L. Golej,1 Andrew T. Bender,2 Joey Liu,3 Willa A. Hsueh,3 Joseph A. Beavo,2 Rosalind A. Coleman,4 and Karin E. Bornfeldt1 Rosiglitazone is an insulin-sensitizing agent that has re- cently been shown to exert beneficial effects on atheroscle- rosis. In addition to peroxisome proliferator–activated ong-chain acyl-CoA synthetases (ACSLs) receptor (PPAR)-␥, rosiglitazone can affect other targets, (E.C.6.2.1.3) catalyze esterification of long-chain such as directly inhibiting recombinant long-chain acyl- fatty acids, mediating the partitioning of fatty CoA synthetase (ACSL)-4 activity. Because it is unknown if acids in mammalian cells. ACSL isoforms ACSL4 is expressed in vascular cells involved in athero- L sclerosis, we investigated the ability of rosiglitazone to (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6) generate inhibit ACSL activity and fatty acid partitioning in human bioactive fatty acyl-CoAs from CoA, ATP, and long-chain and murine arterial smooth muscle cells (SMCs) and mac- (C12–C20) fatty acids (1). ACSLs exhibit different tissue rophages. Human and murine SMCs and human macro- distribution, subcellular localization, fatty acid preference, phages expressed Acsl4, and rosiglitazone inhibited Acsl responsiveness to pharmacological inhibitors, and tran- activity in these cells. Furthermore, rosiglitazone acutely scriptional regulation. Previous studies have characterized inhibited partitioning of fatty acids into phospholipids in these enzymes in different tissues (2); however, expression human SMCs and inhibited fatty acid partitioning into levels and function in vascular cells are unknown. Until diacylglycerol and triacylglycerol in human SMCs and mac- rophages through a PPAR-␥–independent mechanism. Con- recently, inconsistencies in the ACSL nomenclature led to versely, murine macrophages did not express ACSL4, and confusion in the literature. The standard nomenclature rosiglitazone did not inhibit ACSL activity in these cells, now uses ACSL1 and ACSL3–6 for human and Acsl1 and nor did it affect acute fatty acid partitioning into cellular Acsl3–6 for murine members of this family (3). lipids. Thus, rosiglitazone inhibits ACSL activity and fatty Thiazolidinediones (TZDs) are a class of oral insulin- acid partitioning in human and murine SMCs and in human sensitizing agents, extensively used in the treatment of macrophages through a PPAR-␥–independent mechanism type 2 diabetes. Whereas members of this class of agents, likely to be mediated by ACSL4 inhibition. Therefore, such as rosiglitazone and pioglitazone, have been demon- rosiglitazone might alter the biological effects of fatty acids in these cells and in atherosclerosis. Diabetes 56: strated to provide effective glycemic control in patients 1143–1152, 2007 with diabetes (4), their effect on lipid metabolism is less clear (5). Perhaps more interesting are the recent findings that rosiglitazone treatment results in fewer inflammatory cells and increased collagen content in advanced athero- From the 1Department of Pathology, University of Washington School of sclerotic plaques (6,7; rev. in 8), suggesting that rosiglita- Medicine, Seattle, Washington; the 2Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington; the 3Departments of zone might exert a stabilizing effect on atherosclerotic Nutrition and Pediatrics, University of North Carolina, Chapel Hill, North plaques. Carolina; and the 4Division of Endocrinology, Diabetes, and Hypertension, The mechanism of TZD action is mediated largely David E. Geffen School of Medicine, University of California, Los Angeles, ␥ California. through the activation of PPAR- , a member of the super Address correspondence and reprint requests to Dr. Karin E. Bornfeldt, family of ligand-activated nuclear transcription factors. In Department of Pathology, 1959 NE Pacific St., University of Washington, vascular tissue, peroxisome proliferator–activated recep- Seattle, WA 98195-7470. E-mail: [email protected]. Received for publication 24 February 2006 and accepted in revised form 16 tor (PPAR)-␥ is expressed in smooth muscle cells, macro- January 2007. phages, and endothelial cells. TZDs may also exert Published ahead of print at http://diabetes.diabetesjournals.org on 26 Jan- nongenomic effects. For example, rosiglitazone has been uary 2007. DOI: 10.2337/db06-0267. ⌬12,14 shown to activate 5Ј-AMP protein kinase (AMPK) through 15-dPGJ2, 15-deoxy- prostaglandin J2; AA, arachidonic acid; ACSL, long-chain acyl-CoA synthetase; AICAR, 5-aminoimidazole-4-carboxaide 1-␤- a PPAR-␥–independent mechanism (9). With regard to D-ribo-furanoside; DAG, diacylglycerol; hSMC, human aortic smooth muscle Acsl isoforms, in vitro studies of rat recombinant proteins cell; IC50, half-maximal inhibition; OA, oleic acid; SMC, smooth muscle cell; TAG, triacylglycerol; TZD, thiazolidinedione. have demonstrated that TZDs can directly inhibit the © 2007 by the American Diabetes Association. activity of one of the gene products, Acsl4 (10). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance Because of the strong association among increased fatty with 18 U.S.C. Section 1734 solely to indicate this fact. acid load, atherosclerosis, and cardiovascular disease, DIABETES, VOL. 56, APRIL 2007 1143 ROSIGLITAZONE INHIBITS ACSL4 ACTIVITY particularly in the setting of type 2 diabetes, we investi- separated via gel electrophoresis in 2% agarose gels and visualized with gated the ability of rosiglitazone to inhibit ACSL activity ethidium bromide. Murine brain and liver were used as positive controls for the Acsl3, Acsl5, and Acsl6 primers. A human neuroblastoma cell line and acute fatty acid partitioning in smooth muscle cells (SH-SY5Y) was used as a positive control for human ACSL6. (SMCs) and macrophages. We show that human and Western blot analysis of ACSL1 and ACSL4. Lysates were prepared as murine SMCs and human macrophages express a number previously described (12,14,16), separated using 7.5% Tris-glycine SDS-PAGE, of ACSL isoforms and that ACSL4, the rosiglitazone- and transferred to polyvinylidene difluoride membranes. The membranes sensitive isoform, is expressed in these cells. We also were then incubated overnight at 4°C with rabbit polyclonal anti-rat Acsl1 or show that rosiglitazone inhibits ACSL activity and fatty Acsl4 antibodies (1:10,000) generated against the amino acid sequences acid partitioning in human and murine SMCs and in human MEVHELFRYFRMPELIDIR and EIHSMQSVEELGSKPENSSI, respectively ␥ (17). Horseradish peroxidase–linked anti-rabbit secondary antibodies were macrophages through a PPAR- –independent mechanism used at a final dilution of 1:5,000 for1hatroom temperature. Signal detection likely to be mediated by direct inhibition of ACSL4. was performed by enhanced chemiluminescence. Recombinant rat Acsl4 (3 Furthermore, rosiglitazone might exert effects on fatty ␮g) was used as a positive control. Rat liver high-speed supernatant was used acid partitioning in human macrophages that are not as a positive control for ACSL1 expression. Briefly, rat liver was homogenized mimicked by mouse models, because murine macro- (1:3; weight:volume) using a Potter-Elvehjem homogenizer in a 320 mmol/l sucrose buffer containing Tris-HCl, pH 7.4 (20 mmol/l), EDTA (1 mmol/l), phages are devoid of Acsl4. EGTA (1 mmol/l), leupeptin (20 ␮g/ml), benzamidine (1 mmol/l), aprotinin (10 ␮g/ml), pepstatin (5 ␮g/ml), dithiothreitol (1 mmol/l), and phenylmethylsulfo- nyl fluoride (0.1 mmol/l). The homogenate was centrifuged at 7,700g for 2 min. RESEARCH DESIGN AND METHODS The supernatant was then centrifuged at 12,000g for 12 min and then at Oleic acid (OA) (sodium salt) was purchased from Nucheck Prep (Elysian, 177,000g for 1 h. The resulting high-speed supernatant was used for analyses. MN). RPMI-1640, fetal bovine serum, and penicillin/streptomycin were ob- Analysis of ACSL activity. Total acyl-CoA synthetase activity was measured tained from Gibco/Invitrogen (Grand Island, NY). CoA, ATP, essentially fatty via a modified protocol by Kim et al. (10), involving the formation of OA-CoA acid–free BSA, sodium thioglycollate, dithiothreitol, EDTA, EGTA, leupeptin, from OA. Briefly, cells were scraped into ice-cold cell sonication buffer aprotinin, pepstatin, phenylmethylsulfonyl fluoride, and benzamidine were containing 50 mmol/l potassium phosphate (pH 7.4), 10% glycerol, 1 mmol/l obtained from Sigma-Aldrich (St. Louis, MO). Triascin C, T0070907, 5-amino- EDTA, 20 ␮g/ml leupeptin, 5 ␮g/ml pepstatin, 10 ␮g/ml aprotinin, and 5 mmol/l imidazole-4-carboxaide 1-␤-D-ribo-furanoside (AICAR), 15-deoxy-⌬12,14 prosta- benzamidine and sonicated with two bursts of 10-s duration at 50 W. Protein glandin J2 (15-dPGJ2), and Wy14643 were from Biomol Research Laboratories concentration was determined via BCA protein assays (Pierce, Rockford, IL). (Plymouth Meeting, PA). Rosiglitazone was obtained from Alexis Biochemical The lysate (200 ␮g/sample) was then incubated in the presence of increasing (San Diego, CA). Anti-rabbit IgG horseradish peroxidase–linked antibodies, concentrations of rosiglitazone, triacsin C, 15-dPGJ2, AICAR, or Wy16463
Recommended publications
  • Cell Death Via Lipid Peroxidation and Protein Aggregation Diseases
    biology Review Cell Death via Lipid Peroxidation and Protein Aggregation Diseases Katsuya Iuchi * , Tomoka Takai and Hisashi Hisatomi Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, 3-3-1 Kichijojikitamachi, Musashino-shi, Tokyo 180-8633, Japan; [email protected] (T.T.); [email protected] (H.H.) * Correspondence: [email protected] or [email protected]; Tel.: +81-422-37-3523 Simple Summary: It is essential for cellular homeostasis that biomolecules, such as DNA, proteins, and lipids, function properly. Disturbance of redox homeostasis produces aberrant biomolecules, including oxidized lipids and misfolded proteins, which increase in cells. Aberrant biomolecules are removed by excellent cellular clearance systems. However, when excess aberrant biomolecules remain in the cell, they disrupt organelle and cellular functions, leading to cell death. These aberrant molecules aggregate and cause apoptotic and non-apoptotic cell death, leading to various protein aggregation diseases. Thus, we investigated the cell-death cross-linking between lipid peroxidation and protein aggregation. Abstract: Lipid peroxidation of cellular membranes is a complicated cellular event, and it is both the cause and result of various diseases, such as ischemia-reperfusion injury, neurodegenerative diseases, and atherosclerosis. Lipid peroxidation causes non-apoptotic cell death, which is associated with cell fate determination: survival or cell death. During the radical chain reaction of lipid peroxidation, Citation: Iuchi, K.; Takai, T.; various oxidized lipid products accumulate in cells, followed by organelle dysfunction and the Hisatomi, H. Cell Death via Lipid induction of non-apoptotic cell death. Highly reactive oxidized products from unsaturated fatty acids Peroxidation and Protein are detected under pathological conditions.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Loss of Fam60a, a Sin3a Subunit, Results in Embryonic Lethality and Is Associated with Aberrant Methylation at a Subset of Gene
    RESEARCH ARTICLE Loss of Fam60a, a Sin3a subunit, results in embryonic lethality and is associated with aberrant methylation at a subset of gene promoters Ryo Nabeshima1,2, Osamu Nishimura3,4, Takako Maeda1, Natsumi Shimizu2, Takahiro Ide2, Kenta Yashiro1†, Yasuo Sakai1, Chikara Meno1, Mitsutaka Kadota3,4, Hidetaka Shiratori1†, Shigehiro Kuraku3,4*, Hiroshi Hamada1,2* 1Developmental Genetics Group, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; 2Laboratory for Organismal Patterning, RIKEN Center for Developmental Biology, Kobe, Japan; 3Phyloinformatics Unit, RIKEN Center for Life Science Technologies, Kobe, Japan; 4Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan Abstract We have examined the role of Fam60a, a gene highly expressed in embryonic stem cells, in mouse development. Fam60a interacts with components of the Sin3a-Hdac transcriptional corepressor complex, and most Fam60a–/– embryos manifest hypoplasia of visceral organs and die in utero. Fam60a is recruited to the promoter regions of a subset of genes, with the expression of these genes being either up- or down-regulated in Fam60a–/– embryos. The DNA methylation level of the Fam60a target gene Adhfe1 is maintained at embryonic day (E) 7.5 but markedly reduced at –/– *For correspondence: E9.5 in Fam60a embryos, suggesting that DNA demethylation is enhanced in the mutant. [email protected] (SK); Examination of genome-wide DNA methylation identified several differentially methylated regions, [email protected] (HH) which were preferentially hypomethylated, in Fam60a–/– embryos. Our data suggest that Fam60a is †These authors contributed required for proper embryogenesis, at least in part as a result of its regulation of DNA methylation equally to this work at specific gene promoters.
    [Show full text]
  • Mergeomics: Multidimensional Data Integration to Identify Pathogenic Perturbations to Biological Systems
    Shu et al. BMC Genomics (2016) 17:874 DOI 10.1186/s12864-016-3198-9 METHODOLOGY ARTICLE Open Access Mergeomics: multidimensional data integration to identify pathogenic perturbations to biological systems Le Shu1, Yuqi Zhao1, Zeyneb Kurt1, Sean Geoffrey Byars2,3, Taru Tukiainen4, Johannes Kettunen4, Luz D. Orozco5, Matteo Pellegrini5, Aldons J. Lusis6, Samuli Ripatti4, Bin Zhang7, Michael Inouye2,3,8, Ville-Petteri Mäkinen1,9,10,11* and Xia Yang1,12* Abstract Background: Complex diseases are characterized by multiple subtle perturbations to biological processes. New omics platforms can detect these perturbations, but translating the diverse molecular and statistical information into testable mechanistic hypotheses is challenging. Therefore, we set out to create a public tool that integrates these data across multiple datasets, platforms, study designs and species in order to detect the most promising targets for further mechanistic studies. Results: We developed Mergeomics, a computational pipeline consisting of independent modules that 1) leverage multi-omics association data to identify biological processes that are perturbed in disease, and 2) overlay the disease- associated processes onto molecular interaction networks to pinpoint hubs as potential key regulators. Unlike existing tools that are mostly dedicated to specific data type or settings, the Mergeomics pipeline accepts and integrates datasets across platforms, data types and species. We optimized and evaluated the performance of Mergeomics using simulation and multiple independent datasets, and benchmarked the results against alternative methods. We also demonstrate the versatility of Mergeomics in two case studies that include genome-wide, epigenome-wide and transcriptome-wide datasets from human and mouse studies of total cholesterol and fasting glucose.
    [Show full text]
  • Regulation of Mitochondrial and Nonmitochondrial Protein Turnover by the PINK1-Parkin Pathway
    Regulation of mitochondrial and nonmitochondrial protein turnover by the PINK1-Parkin pathway Evelyn S. Vincow A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy University of Washington 2013 Reading Committee: Leo J. Pallanck, Chair Sandra M. Bajjalieh Michael J. MacCoss Program Authorized to Offer Degree: Neurobiology and Behavior © Copyright 2013 Evelyn S. Vincow University of Washington Abstract Regulation of mitochondrial and nonmitochondrial protein turnover by the PINK1-Parkin pathway Evelyn Sandra Vincow Chair of the Supervisory Committee: Associate Professor Leo J. Pallanck Genome Sciences The accumulation of damaged mitochondria has been proposed as a key factor in aging and in the pathogenesis of many common age-related diseases, including Parkinson disease (PD). Recently, in vitro studies of the PD-related proteins Parkin and PINK1 have found that these factors act in a common pathway to promote the selective autophagic degradation of damaged mitochondria (mitophagy). However, whether PINK1 and Parkin promote mitophagy in vivo is unknown. To address this question, I used a proteomic approach in Drosophila to study the effects of null mutations in parkin or PINK1 on mitochondrial protein turnover. The parkin null mutants showed a significant overall slowing of mitochondrial protein turnover, similar to but less severe than the slowing seen in autophagy-deficient Atg7 mutants, consistent with the model that Parkin acts upstream of Atg7 to promote mitophagy. By contrast, the turnover of many mitochondrial respiratory chain (RC) subunits showed greater impairment in parkin than in Atg7 mutants, and RC turnover was also selectively impaired in PINK1 mutants. These findings demonstrate that the PINK1-Parkin pathway promotes mitophagy in vivo and, unexpectedly, also promotes selective turnover of mitochondrial RC components.
    [Show full text]
  • An ACSL4 Hemizygous Intragenic Deletion in a Patient with Childhood Stroke
    Pediatric Neurology 100 (2019) 100e101 Contents lists available at ScienceDirect Pediatric Neurology journal homepage: www.elsevier.com/locate/pnu Clinical Letter An ACSL4 Hemizygous Intragenic Deletion in a Patient With Childhood Stroke Caitlin A. Chang, MD a, Julie Lauzon, MD, MHSc a, b, Adam Kirton, MD, MSc b, c, * Bob Argiropoulos, PhD a, b, d, a Department of Medical Genetics, Alberta Children's Hospital, Calgary, Alberta, Canada b Alberta Children's Hospital Research Institute for Child and Maternal Health, Alberta Children's Hospital, Calgary, Alberta, Canada c Department of Pediatrics and Clinical Neurosciences, Pediatric Neurology, Alberta Children's Hospital, Calgary, Alberta, Canada d Genetic Laboratory Services, Cytogenetics Laboratory, Alberta Children's Hospital, Calgary, Alberta, Canada article info Article history: Received 4 March 2019 Accepted 22 June 2019 Available online 28 June 2019 Keywords: Childhood stroke ACSL4 X-linked intellectual disability Ischemia Developmental delay We describe a male with a maternally inherited 9.75-kb intra- was present. Additional evaluation for stroke risk factors, including genic deletion of ACSL4. Fewer than 25 individuals have been re- lumbar puncture, echocardiogram, and examination of blood for fl ported with ACSL4-related X-linked intellectual disability (XLID). thrombophilia and in ammatory markers were negative. This individual highlights a unique presentation of ACSL4-related At three years, severe communication delays were evident. fi XLID and reveals additional pathways for investigation. Behavioral dif culties included frequent outbursts, irritability, and tantrums. There were no limitations in gross or fine motor func- Patient description tioning. At 4.5 years, he developed seizures characterized by eye deviation and automatisms.
    [Show full text]
  • Supplementary Material
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Neurol Neurosurg Psychiatry Page 1 / 45 SUPPLEMENTARY MATERIAL Appendix A1: Neuropsychological protocol. Appendix A2: Description of the four cases at the transitional stage. Table A1: Clinical status and center proportion in each batch. Table A2: Complete output from EdgeR. Table A3: List of the putative target genes. Table A4: Complete output from DIANA-miRPath v.3. Table A5: Comparison of studies investigating miRNAs from brain samples. Figure A1: Stratified nested cross-validation. Figure A2: Expression heatmap of miRNA signature. Figure A3: Bootstrapped ROC AUC scores. Figure A4: ROC AUC scores with 100 different fold splits. Figure A5: Presymptomatic subjects probability scores. Figure A6: Heatmap of the level of enrichment in KEGG pathways. Kmetzsch V, et al. J Neurol Neurosurg Psychiatry 2021; 92:485–493. doi: 10.1136/jnnp-2020-324647 BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Neurol Neurosurg Psychiatry Appendix A1. Neuropsychological protocol The PREV-DEMALS cognitive evaluation included standardized neuropsychological tests to investigate all cognitive domains, and in particular frontal lobe functions. The scores were provided previously (Bertrand et al., 2018). Briefly, global cognitive efficiency was evaluated by means of Mini-Mental State Examination (MMSE) and Mattis Dementia Rating Scale (MDRS). Frontal executive functions were assessed with Frontal Assessment Battery (FAB), forward and backward digit spans, Trail Making Test part A and B (TMT-A and TMT-B), Wisconsin Card Sorting Test (WCST), and Symbol-Digit Modalities test.
    [Show full text]
  • Identification of ACSL4 As a Biomarker and Contributor of Ferroptosis In
    Identication of ACSL4 as a biomarker and contributor of ferroptosis in clear cell renal cell carcinoma na guo ( [email protected] ) Primary research Keywords: Acyl-CoA synthetase long-chain family member 4 (ACSL4), biomarker, ferroptosis, clear cell renal cell carcinoma (ccRCC) Posted Date: May 15th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-24079/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/17 Abstract Background ACSL4 has been reported to be related to tumor genesis and involved in the processes of ferroptosis. However, the expression levels and prognostic value of ACSL4 in clear cell renal cell carcinoma (ccRCC) remain unclear. Methods The Oncomine and TCGA databases were used to predict the expression of ACSL4 mRNA in ccRCC and its association with ccRCC prognosis. The expression levels of ACSL4 were determined in human RCC tissues by real-time PCR. Kaplan-Meier curves were used to analyze the diagnostic and prognostic signicance of ACSL4 in ccRCC. A ferroptosis inducer (erastin) was used to investigate the effects of ACSL4 on ferroptosis in ccRCC cell lines. Results The expression level of ACSL4 was signicantly down-regulated in ccRCC tissues (P < 0.001), which was consistent with the analysis of the Oncomine and TCGA database. Then, immunohistochemical results demonstrated that the ACSL4 was weak or not detected in ccRCC tissues than that in normal tissues. ACSL4 differential expression level was signicantly related to gender, ccRCC subtypes, nodal invasion, tumor grade and cancer stages (all P < 0.001). Survival analysis revealed that overall survival was favorable in ccRCC patients with ACSL4 high expression (P = 0.014).
    [Show full text]
  • Global Ubiquitylation Analysis of Mitochondria in Primary Neurons
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.01.438131; this version posted April 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Global ubiquitylation analysis of mitochondria in primary neurons identifies physiological Parkin targets following activation of PINK1 Odetta Antico1#, Alban Ordureau2#, Michael Stevens1, Francois Singh1, Marek φ Gierlinski3, Erica Barini1 , Mollie L. Rickwood1, Alan Prescott4, Rachel Toth1, Ian G. Ganley1, J. Wade Harper2*, and Miratul M. K. Muqit1* 1MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom, DD1 5EH, U.K. 2Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA 3Data Analysis Group, Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom, DD1 5EH, U.K. 4Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, United Kingdom, DD1 5EH, U.K #Denotes Equal Contribution φCurrent address: AbbVie Deutschland GmbH & Co, Knollstr, 67061, Ludwigshafen, Germany *Correspondence: [email protected] or [email protected] Keywords Neurons, PINK1, Parkin, ubiquitin, Parkinson’s disease, Mitochondria, Running Title: Ubiquitin analysis of mitochondria in neurons 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.01.438131; this version posted April 1, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • A Unique Retrogene Within a Gene That Has Acquired Multiple Promoters and a Specific Function in Spermatogenesis
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Developmental Biology 304 (2007) 848–859 www.elsevier.com/locate/ydbio Genomes & Developmental Control Utp14b: A unique retrogene within a gene that has acquired multiple promoters and a specific function in spermatogenesis Ming Zhao a,1, Jan Rohozinski b,1, Manju Sharma c, Jun Ju a, Robert E. Braun c, ⁎ Colin E. Bishop b,d, Marvin L. Meistrich a, a Department of Experimental Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Box 066, 1515 Holcombe Blvd, Houston, TX 77030, USA b Department of Obstetrics and Gynecology, Baylor College of Medicine, 1709 Dryden Road, Houston, TX 77030, USA c Department of Genome Sciences, University of Washington School of Medicine, Box 357730, 1705 N.E. Pacific Street, Seattle, WA 98195, USA d Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Received for publication 28 September 2006; revised 9 December 2006; accepted 3 January 2007 Available online 9 January 2007 Abstract The mouse retrogene Utp14b is essential for male fertility, and a mutation in its sequence results in the sterile juvenile spermatogonial depletion (jsd) phenotype. It is a retrotransposed copy of the Utp14a gene, which is located on the X chromosome, and is inserted within an intron of the autosomal acyl-CoA synthetase long-chain family member 3 (Acsl3) gene. To elucidate the roles of the Utp14 genes in normal spermatogenic cell development as a basis for understanding the defects that result in the jsd phenotype, we analyzed the various mRNAs produced from the Utp14b retrogene and their expression in different cell types.
    [Show full text]
  • Elucidating Biological Roles of Novel Murine Genes in Hearing Impairment in Africa
    Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 19 September 2019 doi:10.20944/preprints201909.0222.v1 Review Elucidating Biological Roles of Novel Murine Genes in Hearing Impairment in Africa Oluwafemi Gabriel Oluwole,1* Abdoulaye Yal 1,2, Edmond Wonkam1, Noluthando Manyisa1, Jack Morrice1, Gaston K. Mazanda1 and Ambroise Wonkam1* 1Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, South Africa. 2Department of Neurology, Point G Teaching Hospital, University of Sciences, Techniques and Technology, Bamako, Mali. *Correspondence to: [email protected]; [email protected] Abstract: The prevalence of congenital hearing impairment (HI) is highest in Africa. Estimates evaluated genetic causes to account for 31% of HI cases in Africa, but the identification of associated causative genes mutations have been challenging. In this study, we reviewed the potential roles, in humans, of 38 novel genes identified in a murine study. We gathered information from various genomic annotation databases and performed functional enrichment analysis using online resources i.e. genemania and g.proflier. Results revealed that 27/38 genes are express mostly in the brain, suggesting additional cognitive roles. Indeed, HERC1- R3250X had been associated with intellectual disability in a Moroccan family. A homozygous 216-bp deletion in KLC2 was found in two siblings of Egyptian descent with spastic paraplegia. Up to 27/38 murine genes have link to at least a disease, and the commonest mode of inheritance is autosomal recessive (n=8). Network analysis indicates that 20 other genes have intermediate and biological links to the novel genes, suggesting their possible roles in HI.
    [Show full text]
  • Systematic Name Gene Name Systematic Name Gene Name NM 001710 Complement Factor B(CFB) NM 052831 Solute Carrier Family 18 Member
    Table S1: Genome-wide identification of SGLT2i`s interaction with early inflammatory response in human proximal tubular cells. Systematic Systematic Gene Name Gene Name Name Name solute carrier family 18 member NM_001710 complement factor B(CFB) NM_052831 B1(SLC18B1) heterogeneous nuclear DAZ associated protein NM_031372 NM_170711 ribonucleoprotein D like(HNRNPDL) 1(DAZAP1) NM_014299 bromodomain containing 4(BRD4) NM_001261 cyclin dependent kinase 9(CDK9) cilia and flagella associated protein NM_182628 NM_178835 zinc finger protein 827(ZNF827) 100(CFAP100) NM_017906 PAK1 interacting protein 1(PAK1IP1) NM_024015 homeobox B4(HOXB4) family with sequence similarity 167 ankyrin repeat and LEM domain NM_053279 NM_015114 member A(FAM167A) containing 2(ANKLE2) small cell adhesion ARP3 actin related protein 3 NM_001031628 NM_005721 glycoprotein(SMAGP) homolog(ACTR3) TRAF3 interacting protein actin related protein 2/3 complex NM_147686 NM_005720 2(TRAF3IP2) subunit 1B(ARPC1B) basic leucine zipper ATF-like cAMP responsive element binding NM_018664 NM_182898 transcription factor 3(BATF3) protein 5(CREB5) zinc finger CCCH-type containing activation induced cytidine NM_025079 NM_020661 12A(ZC3H12A) deaminase(AICDA) C-X-C motif chemokine ligand DENN domain containing NM_001511 NM_015213 1(CXCL1) 5A(DENND5A) NM_025072 prostaglandin E synthase 2(PTGES2) NM_004665 vanin 2(VNN2) superoxide dismutase 2, mitochondrial ribosomal protein NM_001024465 NM_016070 mitochondrial(SOD2) S23(MRPS23) jumonji and AT-rich interaction NM_033199 urocortin 2(UCN2) NM_004973
    [Show full text]