1
|
Challa NL, Sarkar A, Kapettu S, Phanithi PB, Chakrabarti P, Parsa KVL, Misra P. TGS1/PIMT regulates pro-inflammatory macrophage mediated paracrine insulin resistance: Crosstalk between macrophages and skeletal muscle cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166878. [PMID: 37673359 DOI: 10.1016/j.bbadis.2023.166878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Macrophage-driven chronic low-grade inflammatory response is intimately associated with pathogenesis of insulin resistance and type 2 diabetes (T2D). However, the molecular basis for skewing of pro-inflammatory macrophage is still elusive. Here, we describe the mechanism and significance of TGS1/PIMT (PRIP-Interacting protein with Methyl Transferase domain) in regulating macrophage activation and polarization and its impact on the development of insulin resistance in skeletal muscle cells. We show altered expression of TGS1 in M1 polarized cultured macrophages, bone marrow-derived (BMDM) and adipose tissue macrophages. Moreover, in High Fat Diet (HFD)-fed mice enhanced TGS1 expression is predominantly localized to the nucleus of adipose tissue macrophages suggesting its potential functional role. Gain and loss of TGS1 expression in macrophage further established its role in the secretion of pro-inflammatory mediators. Mechanistically, TGS1 controls the transcription of numerous genes linked to inflammation by forming a complex with Histone Acetyl Transferase (HAT)-containing transcriptional co-activators CBP and p300. Functionally, TGS1 mediated macrophage inflammatory response induces the development of insulin resistance in skeletal muscle cells and adipocytes. Our findings thus demonstrate an unexpected contribution of TGS1 in the regulation of macrophage mediated inflammation and insulin resistance highlighting that TGS1 antagonism could be a promising therapeutic target for the management of inflammation and insulin resistance in T2D.
Collapse
Affiliation(s)
- Naga Lakshmi Challa
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life sciences, University of Hyderabad Campus, Hyderabad 500046, Telangana, India; Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India; Registered as a PhD student with MAHE, Manipal, India
| | - Ankita Sarkar
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India
| | - Satyamoorthy Kapettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Prakash Babu Phanithi
- Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
| | - Kishore V L Parsa
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life sciences, University of Hyderabad Campus, Hyderabad 500046, Telangana, India.
| | - Parimal Misra
- Centre for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life sciences, University of Hyderabad Campus, Hyderabad 500046, Telangana, India.
| |
Collapse
|
2
|
Zhong J, Yuan C, Liu L, Du Y, Hui Y, Chen Z, Diao C, Yang R, Liu G, Liu X. PCMT1 regulates the migration, invasion, and apoptosis of prostate cancer through modulating the PI3K/AKT/GSK-3β pathway. Aging (Albany NY) 2023; 15:11654-11671. [PMID: 37899170 PMCID: PMC10637816 DOI: 10.18632/aging.205152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023]
Abstract
Protein L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) is a repair enzyme that catalyzes the conversion of isomerized aspartic acid (iso-Asp) residues into their normal structure, thereby restoring the configuration and function of proteins. Studies have shown that PCMT1 is overexpressed in several tumors and affects patients' prognosis. However, there are few reports on the role of PCMT1 in prostate cancer (PCa). In the present research, with the assistance of The Cancer Genome Atlas Program (TCGA) database, we found that PCMT1 was overexpressed in PCa tissues. The results of quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blot and immunohistochemistry staining also showed that PCMT1 expression was significantly increased in PCa tissues and cell lines. In PCa clinical samples, PCMT1 expression was closely related to Gleason score, clinical stage, lymph node metastasis and bone metastasis. The experiments of overexpression and knockdown of PCMT1 in vitro or in vivo showed that PCMT1 can significantly promote the proliferation, migration and invasion of PCa cells, inhibit cell apoptosis, and promote the growth of PCa. We furthermore confirmed that PCMT1 regulated the migration, invasion and apoptosis of PCa cells by modulating the phosphatidylinositol 3-kinase/AKT kinase/glycogen-synthase kinase-3β (PI3K/AKT/GSK-3β) signaling pathway. Collectively, PCMT1 plays a cancer-facilitative role in PCa by promoting the proliferation, migration and invasion of PCa cells, and inhibiting apoptosis. Therefore, PCMT1 is considered to represent a novel target for treating PCa.
Collapse
Affiliation(s)
- Jiacheng Zhong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Yuan
- Department of Urology, Jingzhou Central Hospital, Jingzhou 434020, China
| | - Lin Liu
- Department of Emergency, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yang Du
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yumin Hui
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Changhui Diao
- Department of Urology, The First People’s Hospital of Shangqiu City, Shangqiu 476100, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guiyong Liu
- Department of Urology, Qianjiang Central Hospital, Qianjiang 433100, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
3
|
Siena LA, Michaud C, Selles B, Vega JM, Pessino SC, Ingouff M, Ortiz JPA, Leblanc O. TRIMETHYLGUANOSINE SYNTHASE1 mutations decanalize female germline development in Arabidopsis. THE NEW PHYTOLOGIST 2023; 240:597-612. [PMID: 37548040 DOI: 10.1111/nph.19179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/14/2023] [Indexed: 08/08/2023]
Abstract
Here, we report the characterization of a plant RNA methyltransferase, orthologous to yeast trimethylguanosine synthase1 (Tgs1p) and whose downregulation was associated with apomixis in Paspalum grasses. Using phylogenetic analyses and yeast complementation, we determined that land plant genomes all encode a conserved, specific TGS1 protein. Next, we studied the role of TGS1 in female reproduction using reporter lines and loss-of-function mutants in Arabidopsis thaliana. pAtTGS1:AtTGS1 reporters showed a dynamic expression pattern. They were highly active in the placenta and ovule primordia at emergence but, subsequently, showed weak signals in the nucellus. Although expressed throughout gametophyte development, activity became restricted to the female gamete and was also detected after fertilization during embryogenesis. TGS1 depletion altered the specification of the precursor cells that give rise to the female gametophytic generation and to the sporophyte, resulting in the formation of a functional aposporous-like lineage. Our results indicate that TGS1 participates in the mechanisms restricting cell fate acquisition to a single cell at critical transitions throughout the female reproductive lineage and, thus, expand our current knowledge of the mechanisms governing female reproductive fate in plants.
Collapse
Affiliation(s)
- Lorena A Siena
- Instituto de Investigaciones en Ciencias Agrarias de Rosario, CONICET-Universidad Nacional de Rosario, S2125ZAA, Zavalla, Argentina
| | | | - Benjamin Selles
- DIADE, Univ Montpellier, IRD, CIRAD, 34394, Montpellier, France
| | - Juan Manuel Vega
- Instituto de Investigaciones en Ciencias Agrarias de Rosario, CONICET-Universidad Nacional de Rosario, S2125ZAA, Zavalla, Argentina
| | - Silvina C Pessino
- Instituto de Investigaciones en Ciencias Agrarias de Rosario, CONICET-Universidad Nacional de Rosario, S2125ZAA, Zavalla, Argentina
| | - Mathieu Ingouff
- DIADE, Univ Montpellier, IRD, CIRAD, 34394, Montpellier, France
| | - Juan Pablo A Ortiz
- Instituto de Investigaciones en Ciencias Agrarias de Rosario, CONICET-Universidad Nacional de Rosario, S2125ZAA, Zavalla, Argentina
| | - Olivier Leblanc
- DIADE, Univ Montpellier, IRD, CIRAD, 34394, Montpellier, France
| |
Collapse
|
4
|
Sharma R, Maity SK, Chakrabarti P, Katika MR, Kapettu S, Parsa KVL, Misra P. PIMT Controls Insulin Synthesis and Secretion through PDX1. Int J Mol Sci 2023; 24:ijms24098084. [PMID: 37175791 PMCID: PMC10179560 DOI: 10.3390/ijms24098084] [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: 01/06/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic beta cell function is an important component of glucose homeostasis. Here, we investigated the function of PIMT (PRIP-interacting protein with methyl transferase domain), a transcriptional co-activator binding protein, in the pancreatic beta cells. We observed that the protein levels of PIMT, along with key beta cell markers such as PDX1 (pancreatic and duodenal homeobox 1) and MafA (MAF bZIP transcription factor A), were reduced in the beta cells exposed to hyperglycemic and hyperlipidemic conditions. Consistently, PIMT levels were reduced in the pancreatic islets isolated from high fat diet (HFD)-fed mice. The RNA sequencing analysis of PIMT knockdown beta cells identified that the expression of key genes involved in insulin secretory pathway, Ins1 (insulin 1), Ins2 (insulin 2), Kcnj11 (potassium inwardly-rectifying channel, subfamily J, member 11), Kcnn1 (potassium calcium-activated channel subfamily N member 1), Rab3a (member RAS oncogene family), Gnas (GNAS complex locus), Syt13 (synaptotagmin 13), Pax6 (paired box 6), Klf11 (Kruppel-Like Factor 11), and Nr4a1 (nuclear receptor subfamily 4, group A, member 1) was attenuated due to PIMT depletion. PIMT ablation in the pancreatic beta cells and in the rat pancreatic islets led to decreased protein levels of PDX1 and MafA, resulting in the reduction in glucose-stimulated insulin secretion (GSIS). The results from the immunoprecipitation and ChIP experiments revealed the interaction of PIMT with PDX1 and MafA, and its recruitment to the insulin promoter, respectively. Importantly, PIMT ablation in beta cells resulted in the nuclear translocation of insulin. Surprisingly, forced expression of PIMT in beta cells abrogated GSIS, while Ins1 and Ins2 transcript levels were subtly enhanced. On the other hand, the expression of genes, PRIP/Asc2/Ncoa6 (nuclear receptor coactivator 6), Pax6, Kcnj11, Syt13, Stxbp1 (syntaxin binding protein 1), and Snap25 (synaptosome associated protein 25) associated with insulin secretion, was significantly reduced, providing an explanation for the decreased GSIS upon PIMT overexpression. Our findings highlight the importance of PIMT in the regulation of insulin synthesis and secretion in beta cells.
Collapse
Affiliation(s)
- Rahul Sharma
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| | - Sujay K Maity
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Madhumohan R Katika
- Central Research Lab Mobile Virology Research & Diagnostics BSL3 Lab, ESIC Medical College and Hospital, Hyderabad 500038, India
| | - Satyamoorthy Kapettu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Kishore V L Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad 500046, India
| |
Collapse
|
5
|
Kapadia B, Behera S, Kumar ST, Shah T, Edwin RK, Babu PP, Chakrabarti P, Parsa KV, Misra P. PIMT regulates hepatic gluconeogenesis in mice. iScience 2023; 26:106120. [PMID: 36866247 PMCID: PMC9972567 DOI: 10.1016/j.isci.2023.106120] [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: 08/01/2022] [Revised: 11/29/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The physiological and metabolic functions of PIMT/TGS1, a third-generation transcriptional apparatus protein, in glucose homeostasis sustenance are unclear. Here, we observed that the expression of PIMT was upregulated in the livers of short-term fasted and obese mice. Lentiviruses expressing Tgs1-specific shRNA or cDNA were injected into wild-type mice. Gene expression, hepatic glucose output, glucose tolerance, and insulin sensitivity were evaluated in mice and primary hepatocytes. Genetic modulation of PIMT exerted a direct positive impact on the gluconeogenic gene expression program and hepatic glucose output. Molecular studies utilizing cultured cells, in vivo models, genetic manipulation, and PKA pharmacological inhibition establish that PKA regulates PIMT at post-transcriptional/translational and post-translational levels. PKA enhanced 3'UTR-mediated translation of TGS1 mRNA and phosphorylated PIMT at Ser656, increasing Ep300-mediated gluconeogenic transcriptional activity. The PKA-PIMT-Ep300 signaling module and associated PIMT regulation may serve as a key driver of gluconeogenesis, positioning PIMT as a critical hepatic glucose sensor.
Collapse
Affiliation(s)
- Bandish Kapadia
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad, TG 500046, India
| | - Soma Behera
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad, TG 500046, India
| | - Sireesh T. Kumar
- Department of Biotechnology, University of Hyderabad, Hyderabad 500046, India
| | - Tapan Shah
- Department of Biochemistry, Saurashtra University, Rajkot 360005, India
| | - Rebecca Kristina Edwin
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad, TG 500046, India
| | | | | | - Kishore V.L. Parsa
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad, TG 500046, India,Corresponding author
| | - Parimal Misra
- Center for Innovation in Molecular and Pharmaceutical Sciences, Dr. Reddy’s Institute of Life Sciences (DRILS), University of Hyderabad Campus, Hyderabad, TG 500046, India,Corresponding author
| |
Collapse
|
6
|
Spotting the Targets of the Apospory Controller TGS1 in Paspalum notatum. PLANTS 2022; 11:plants11151929. [PMID: 35893633 PMCID: PMC9332697 DOI: 10.3390/plants11151929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022]
Abstract
Sexuality and apomixis are interconnected plant reproductive routes possibly behaving as polyphenic traits under the influence of the environment. In the subtropical grass Paspalum notatum, one of the controllers of apospory, a main component of gametophytic apomixis reproduction, is TRIMETHYLGUANOSINE SYNTHASE 1 (TGS1), a multifunctional gene previously associated with RNA cleavage regulation (including mRNA splicing as well as rRNA and miRNA processing), transcriptional modulation and the establishment of heterochromatin. In particular, the downregulation of TGS1 induces a sexuality decline and the emergence of aposporous-like embryo sacs. The present work was aimed at identifying TGS1 target RNAs expressed during reproductive development of Paspalum notatum. First, we mined available RNA databases originated from spikelets of sexual and apomictic plants, which naturally display a contrasting TGS1 representation, to identify differentially expressed mRNA splice variants and miRNAs. Then, the role of TGS1 in the generation of these particular molecules was investigated in antisense tgs1 sexual lines. We found that CHLOROPHYLL A-B BINDING PROTEIN 1B-21 (LHC Ib-21, a component of the chloroplast light harvesting complex), QUI-GON JINN (QGJ, encoding a MAP3K previously associated with apomixis) and miR2275 (a meiotic 24-nt phasi-RNAs producer) are directly or indirectly targeted by TGS1. Our results point to a coordinated control exercised by signal transduction and siRNA machineries to induce the transition from sexuality to apomixis.
Collapse
|
7
|
PIMT/TGS1: an evolving metabolic molecular switch with conserved methyl transferase activity. Drug Discov Today 2022; 27:2386-2393. [DOI: 10.1016/j.drudis.2022.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/01/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022]
|
8
|
Dixit G, Prabhu A. The pleiotropic peroxisome proliferator activated receptors: Regulation and therapeutics. Exp Mol Pathol 2021; 124:104723. [PMID: 34822814 DOI: 10.1016/j.yexmp.2021.104723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The Peroxisome proliferator-activated receptors (PPARs) are key regulators of metabolic events in our body. Owing to their implication in maintenance of homeostasis, both PPAR agonists and antagonists assume therapeutic significance. Understanding the molecular mechanisms of each of the PPAR isotypes in the healthy body and during disease is crucial to exploiting their full therapeutic potential. This article is an attempt to present a rational analysis of the multifaceted therapeutic effects and underlying mechanisms of isotype-specific PPAR agonists, dual PPAR agonists, pan PPAR agonists as well as PPAR antagonists. A holistic understanding of the mechanistic dimensions of these key metabolic regulators will guide future efforts to identify novel molecules in the realm of metabolic, inflammatory and immunotherapeutic diseases.
Collapse
Affiliation(s)
- Gargi Dixit
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
9
|
Cheng L, Zhang Y, Zhang Y, Chen T, Xu YZ, Rong YS. Loss of the RNA trimethylguanosine cap is compatible with nuclear accumulation of spliceosomal snRNAs but not pre-mRNA splicing or snRNA processing during animal development. PLoS Genet 2020; 16:e1009098. [PMID: 33085660 PMCID: PMC7605716 DOI: 10.1371/journal.pgen.1009098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 11/02/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022] Open
Abstract
The 2,2,7-trimethylguanosine (TMG) cap is one of the first identified modifications on eukaryotic RNAs. TMG, synthesized by the conserved Tgs1 enzyme, is abundantly present on snRNAs essential for pre-mRNA splicing. Results from ex vivo experiments in vertebrate cells suggested that TMG ensures nuclear localization of snRNAs. Functional studies of TMG using tgs1 mutations in unicellular organisms yield results inconsistent with TMG being indispensable for either nuclear import or splicing. Utilizing a hypomorphic tgs1 mutation in Drosophila, we show that TMG reduction impairs germline development by disrupting the processing, particularly of introns with smaller sizes and weaker splice sites. Unexpectedly, loss of TMG does not disrupt snRNAs localization to the nucleus, disputing an essential role of TMG in snRNA transport. Tgs1 loss also leads to defective 3' processing of snRNAs. Remarkably, stronger tgs1 mutations cause lethality without severely disrupting splicing, likely due to the preponderance of TMG-capped snRNPs. Tgs1, a predominantly nucleolar protein in Drosophila, likely carries out splicing-independent functions indispensable for animal development. Taken together, our results suggest that nuclear import is not a conserved function of TMG. As a distinctive structure on RNA, particularly non-coding RNA, we suggest that TMG prevents spurious interactions detrimental to the function of RNAs that it modifies.
Collapse
Affiliation(s)
- Lin Cheng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Hengyang College of Medicine, University of South China, Hengyang, China
| | - Yu Zhang
- Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Yi Zhang
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, Bethesda, United States of America
| | - Tao Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Hengyang College of Medicine, University of South China, Hengyang, China
| | - Yong-Zhen Xu
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Yikang S. Rong
- Hengyang College of Medicine, University of South China, Hengyang, China
- * E-mail:
| |
Collapse
|
10
|
Diversity and Regulation of S-Adenosylmethionine Dependent Methyltransferases in the Anhydrobiotic Midge. INSECTS 2020; 11:insects11090634. [PMID: 32947792 PMCID: PMC7565475 DOI: 10.3390/insects11090634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 01/23/2023]
Abstract
Multiple co-localized paralogs of genes in Polypedilum vanderplanki's genome have strong transcriptional response to dehydration and considered to be a part of adaptation machinery at the larvae stage. One group of such genes represented by L-isoaspartate O-methyltransferases (PIMT). In order to highlight specific role of PIMT paralogization in desiccation tolerance of the larvae we annotated and compared S-adenosylmethionine (SAM) dependent methyltransferases of four insect species. From another side we applied co-expression analysis in desiccation/rehydration time course and showed that PIMT coding genes could be separated into five clusters by expression profile. We found that among Polypedilum vanderplanki's PIMTs only PIMT1 and PIMT2 have enzymatic activity in normal physiological conditions. From in silico analysis of the protein structures we found two highly variable regions outside of the active center, but also amino acid substitutions which may affect SAM stabilization. Overall, in this study we demonstrated features of Polypedilum vanderplanki's PIMT coding paralogs related to different roles in desiccation tolerance of the larvae. Our results also suggest a role of different SAM-methyltransferases in the adaptation, including GSMT, JHAMT, and candidates from other classes, which could be considered in future studies.
Collapse
|
11
|
Jia G, Yang H, Diao Z, Liu Y, Sun C. Neural stem cell conditioned medium alleviates Aβ 25-35 damage to SH-SY5Y cells through the PCMT1/MST1 pathway. Eur J Histochem 2020; 64:3135. [PMID: 32705859 PMCID: PMC7388643 DOI: 10.4081/ejh.2020.3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/29/2020] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disease. Accumulating evidence suggests that protein isoaspartate methyltransferase 1 (PCMT1) is highly expressed in brain tissue (substantia nigra, blue plaque, paraventricular nucleus). In this study, we investigated the effect of neural stem cell conditioned medium alleviates Aβ25-35 damage to SH-SY5Y cells by PCMT1/MST1 pathway. Results demonstrated that Aβ25-35 significantly decreased the cell viability in time and dose dependent manner. However, Neural stem cell-complete medium (NSC-CPM) or NSC-CDM had inhibitory effect on toxicity when fibrillation of Aβ25-35 occurred in their presence and NSC-CDM had a better inhibitor result. An increase of the PCMT1 expression levels was found in Aβ25-35 + NSC-CDM group. sh-PCMT1 significantly reduced the PCMT1, the cell viability and inhibited the protective effect; induced apoptosis and increased the expression of p-MST1. Overexpression of PCMT1 group reversed the effect of Aβ25-35 inhibited the cell viability and Aβ25-35 induced the apoptosis. In conclusion, NSC-CDM corrects the damage of Aβ25-35 to cells by increasing PCMT1, reducing MST phosphorylation.
Collapse
Affiliation(s)
- Guoyong Jia
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Hongna Yang
- Department of Critical-care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Zengyan Diao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Ying Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Congcong Sun
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| |
Collapse
|
12
|
Colono C, Ortiz JPA, Permingeat HR, Souza Canada ED, Siena LA, Spoto N, Galdeano F, Espinoza F, Leblanc O, Pessino SC. A Plant-Specific TGS1 Homolog Influences Gametophyte Development in Sexual Tetraploid Paspalum notatum Ovules. FRONTIERS IN PLANT SCIENCE 2019; 10:1566. [PMID: 31850040 PMCID: PMC6895069 DOI: 10.3389/fpls.2019.01566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 11/08/2019] [Indexed: 05/04/2023]
Abstract
Aposporous apomictic plants form clonal maternal seeds by inducing the emergence of non-reduced (2n) embryo sacs in the ovule nucellus and the development of embryos by parthenogenesis. In previous work, we reported a plant-specific TRIMETHYLGUANOSINE SYNTHASE 1 (TGS1) gene (PN_TGS1-like) showing expression levels positively correlated with sexuality rates in facultative apomictic Paspalum notatum. PN_ TGS1-like displayed contrasting in situ hybridization patterns in apomictic and sexual plant ovules from premeiosis to anthesis. Here we transformed sexual P. notatum with a TGS1-like antisense construction under a constitutive promoter, in order to produce lines with reduced transcript representation. Antisense plants developed prominent trichomes on the adaxial leaf surface, a trait absent from control genotypes. Reproductive development analysis revealed occasional formation of twin ovules. While control individuals typically displayed a single meiotic embryo sac per ovule, antisense lines showed 12.93-15.79% of ovules bearing extra nuclei, which can be assigned to aposporous-like embryo sacs (AES-like) or, alternatively, to gametophytes with a misguided cell fate development. Moreover, around 8.42-9.52% of ovules showed what looked like a combination of meiotic and aposporous-like sacs. Besides, 32.5% of ovules at early developmental stages displayed nucellar cells with prominent nuclei resembling apospory initials (AIs), which surrounded the megaspore mother cell (MMC) or the MMC-derived meiotic products. Two or more concurrent meiosis events were never detected, which suggest a non-reduced nature for the extra nuclei observed in the mature ovules, unless they were generated by proliferation and misguided differentiation of the legitimate meiotic products. The antisense lines produced a similar amount of viable even-sized pollen with respect to control genotypes, and formed an equivalent full seed set (∼9% of total seeds) after self-pollination. Flow cytometry analyses of caryopses derived from antisense lines revealed that all full seeds had originated from meiotic embryo sacs (i.e. by sexuality). A reduction of 25.55% in the germination percentage was detected when comparing antisense lines with controls. Our results indicate that PN_ TGS1-like influences ovule, gametophyte and possibly embryo development.
Collapse
Affiliation(s)
- Carolina Colono
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Juan Pablo A. Ortiz
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Hugo R. Permingeat
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | | | - Lorena A. Siena
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Nicolás Spoto
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Florencia Galdeano
- Genetics Laboratory, IBONE, CONICET, Universidad Nacional del Nordeste, Corrientes, Argentina
| | - Francisco Espinoza
- Genetics Laboratory, IBONE, CONICET, Universidad Nacional del Nordeste, Corrientes, Argentina
| | | | - Silvina C. Pessino
- Molecular Biology Laboratory, IICAR, CONICET, Universidad Nacional de Rosario, Rosario, Argentina
- *Correspondence: Silvina C. Pessino,
| |
Collapse
|
13
|
Jia Y, Liu N, Viswakarma N, Sun R, Schipma MJ, Shang M, Thorp EB, Kanwar YS, Thimmapaya B, Reddy JK. PIMT/NCOA6IP Deletion in the Mouse Heart Causes Delayed Cardiomyopathy Attributable to Perturbation in Energy Metabolism. Int J Mol Sci 2018; 19:ijms19051485. [PMID: 29772707 PMCID: PMC5983783 DOI: 10.3390/ijms19051485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023] Open
Abstract
PIMT/NCOA6IP, a transcriptional coactivator PRIP/NCOA6 binding protein, enhances nuclear receptor transcriptional activity. Germline disruption of PIMT results in early embryonic lethality due to impairment of development around blastocyst and uterine implantation stages. We now generated mice with Cre-mediated cardiac-specific deletion of PIMT (csPIMT−/−) in adult mice. These mice manifest enlargement of heart, with nearly 100% mortality by 7.5 months of age due to dilated cardiomyopathy. Significant reductions in the expression of genes (i) pertaining to mitochondrial respiratory chain complexes I to IV; (ii) calcium cycling cardiac muscle contraction (Atp2a1, Atp2a2, Ryr2); and (iii) nuclear receptor PPAR- regulated genes involved in glucose and fatty acid energy metabolism were found in csPIMT−/− mouse heart. Elevated levels of Nppa and Nppb mRNAs were noted in csPIMT−/− heart indicative of myocardial damage. These hearts revealed increased reparative fibrosis associated with enhanced expression of Tgfβ2 and Ctgf. Furthermore, cardiac-specific deletion of PIMT in adult mice, using tamoxifen-inducible Cre-approach (TmcsPIMT−/−), results in the development of cardiomyopathy. Thus, cumulative evidence suggests that PIMT functions in cardiac energy metabolism by interacting with nuclear receptor coactivators and this property could be useful in the management of heart failure.
Collapse
Affiliation(s)
- Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Ruya Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Mathew J Schipma
- Next Generation Sequencing Core Facility, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Meng Shang
- Feinberg Cardiovascular Research Institute and Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Yashpal S Kanwar
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Janardan K Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
14
|
Ochogavía A, Galla G, Seijo JG, González AM, Bellucci M, Pupilli F, Barcaccia G, Albertini E, Pessino S. Structure, target-specificity and expression of PN_LNC_N13, a long non-coding RNA differentially expressed in apomictic and sexual Paspalum notatum. PLANT MOLECULAR BIOLOGY 2018; 96:53-67. [PMID: 29119346 PMCID: PMC5778186 DOI: 10.1007/s11103-017-0679-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/29/2017] [Indexed: 05/03/2023]
Abstract
KEY MESSAGE: ncRNA PN_LNC_N13 shows contrasting expression in reproductive organs of sexual and apomictic Paspalum notatum genotypes. Apomictic plants set genetically maternal seeds whose embryos derive by parthenogenesis from unreduced egg cells, giving rise to clonal offspring. Several Paspalum notatum apomixis related genes were identified in prior work by comparative transcriptome analyses. Here, one of these candidates (namely N13) was characterized. N13 belongs to a Paspalum gene family including 30-60 members, of which at least eight are expressed at moderate levels in florets. The sequences of these genes show no functional ORFs, but include segments of different protein coding genes. Particularly, N13 shows partial identity to maize gene BT068773 (RESPONSE REGULATOR 6). Secondary structure predictions as well as mature miRNA and target cleavage detection suggested that N13 is not a miRNA precursor. Moreover, N13 family members produce abundant 24-nucleotide small RNAs along extensive parts of their sequences. Surveys in the GREENC and CANTATA databases indicated similarity with plant long non-coding RNAs (lncRNAs) involved in splicing regulation; consequently, N13 was renamed as PN_LNC_N13. The Paspalum BT068773 predicted ortholog (N13TAR) originates floral transcript variants shorter than the canonical maize isoform and with possible structural differences between the apomictic and sexual types. PN_LNC_N13 is expressed only in apomictic plants and displays quantitative representation variation across reproductive developmental stages. However, PN_LNC_N13-like homologs and/or its derived sRNAs showed overall a higher representation in ovules of sexual plants at late premeiosis. Our results suggest the existence of a whole family of N13-like lncRNAs possibly involved in splicing regulation, with some members characterized by differential activity across reproductive types.
Collapse
Affiliation(s)
- Ana Ochogavía
- Instituto de Investigaciones en Ciencias Agrarias de Rosario (IICAR-CONICET), Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, S2125ZAA, Zavalla, Provincia De Santa Fe, Argentina
| | - Giulio Galla
- Laboratory of Genetics and Genomics, DAFNAE, University of Padova, Campus of Agripolis, Viale dell'Università 16, 35020, Legnaro, Italy
| | - José Guillermo Seijo
- Instituto de Botánica del Nordeste (IBONE-CONICET), Facultad de Ciencias Exactas y Naturales y Agrimensura, Facultad de Ciencias Agrarias, Universidad Nacional del Nordeste, Sargento Cabral 2131, 3400, Corrientes, Argentina
| | - Ana María González
- Instituto de Botánica del Nordeste (IBONE-CONICET), Facultad de Ciencias Exactas y Naturales y Agrimensura, Facultad de Ciencias Agrarias, Universidad Nacional del Nordeste, Sargento Cabral 2131, 3400, Corrientes, Argentina
| | - Michele Bellucci
- Institute of Biosciences and Bioresources, Research Division of Perugia, National Research Council (CNR), via della Madonna Alta 130, 06128, Perugia, Italy
| | - Fulvio Pupilli
- Institute of Biosciences and Bioresources, Research Division of Perugia, National Research Council (CNR), via della Madonna Alta 130, 06128, Perugia, Italy
| | - Gianni Barcaccia
- Laboratory of Genetics and Genomics, DAFNAE, University of Padova, Campus of Agripolis, Viale dell'Università 16, 35020, Legnaro, Italy
| | - Emidio Albertini
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Borgo XX Giugno 74, 06121, Perugia, Italy
| | - Silvina Pessino
- Instituto de Investigaciones en Ciencias Agrarias de Rosario (IICAR-CONICET), Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, S2125ZAA, Zavalla, Provincia De Santa Fe, Argentina.
| |
Collapse
|
15
|
Gao J, Wallis JG, Jewell JB, Browse J. Trimethylguanosine Synthase1 (TGS1) Is Essential for Chilling Tolerance. PLANT PHYSIOLOGY 2017; 174:1713-1727. [PMID: 28495891 PMCID: PMC5490903 DOI: 10.1104/pp.17.00340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/09/2017] [Indexed: 05/20/2023]
Abstract
Chilling stress is a major factor limiting plant development and crop productivity. Because the plant response to chilling is so complex, we are far from understanding the genes important in the response to chilling. To identify new genes important in chilling tolerance, we conducted a novel mutant screen, combining a confirmed SALK T-DNA insertion collection with traditional forward genetics. We screened a pool of more than 3700 confirmed homozygous SALK T-DNA insertion lines for visible defects under prolonged growth at 5°C. Of the chilling-sensitive mutants we observed, mutations at one locus were characterized in detail. This gene, At1g45231, encodes an Arabidopsis (Arabidopsis thaliana) trimethylguanosine synthase (TGS1), previously uncharacterized in the plant kingdom. We confirmed that Arabidopsis TGS1 is a functional ortholog of other trimethylguanosine synthases based both on its in vitro methyltransferase activity and on its ability to rescue the cold-growth inhibition of a Saccharomyces cerevisiae tgs1Δ mutant in vivo. While tgs1 mutant plants grew normally at 22°C, their vegetative and reproductive growth was severely compromised under chilling conditions. When we transgenically expressed TGS1 in the mutant plants, the chilling-sensitive phenotype was relieved, demonstrating that TGS1 is required for chilling tolerance.
Collapse
Affiliation(s)
- Jinpeng Gao
- Institute of Biological Chemistry, Clark Hall, Washington State University, Pullman, Washington 99164-6340
| | - James G Wallis
- Institute of Biological Chemistry, Clark Hall, Washington State University, Pullman, Washington 99164-6340
| | - Jeremy B Jewell
- Institute of Biological Chemistry, Clark Hall, Washington State University, Pullman, Washington 99164-6340
| | - John Browse
- Institute of Biological Chemistry, Clark Hall, Washington State University, Pullman, Washington 99164-6340
| |
Collapse
|
16
|
Olivares AM, Moreno-Ramos OA, Haider NB. Role of Nuclear Receptors in Central Nervous System Development and Associated Diseases. J Exp Neurosci 2016; 9:93-121. [PMID: 27168725 PMCID: PMC4859451 DOI: 10.4137/jen.s25480] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 11/13/2022] Open
Abstract
The nuclear hormone receptor (NHR) superfamily is composed of a wide range of receptors involved in a myriad of important biological processes, including development, growth, metabolism, and maintenance. Regulation of such wide variety of functions requires a complex system of gene regulation that includes interaction with transcription factors, chromatin-modifying complex, and the proper recognition of ligands. NHRs are able to coordinate the expression of genes in numerous pathways simultaneously. This review focuses on the role of nuclear receptors in the central nervous system and, in particular, their role in regulating the proper development and function of the brain and the eye. In addition, the review highlights the impact of mutations in NHRs on a spectrum of human diseases from autism to retinal degeneration.
Collapse
Affiliation(s)
- Ana Maria Olivares
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Oscar Andrés Moreno-Ramos
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Neena B Haider
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Tseng CK, Wang HF, Burns A, Schroeder M, Gaspari M, Baumann P. Human Telomerase RNA Processing and Quality Control. Cell Rep 2015; 13:2232-43. [DOI: 10.1016/j.celrep.2015.10.075] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 10/02/2015] [Accepted: 10/27/2015] [Indexed: 12/11/2022] Open
|
18
|
Co-activator binding protein PIMT mediates TNF-α induced insulin resistance in skeletal muscle via the transcriptional down-regulation of MEF2A and GLUT4. Sci Rep 2015; 5:15197. [PMID: 26468734 PMCID: PMC4606566 DOI: 10.1038/srep15197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/21/2015] [Indexed: 12/18/2022] Open
Abstract
The mechanisms underlying inflammation induced insulin resistance are poorly understood. Here, we report that the expression of PIMT, a transcriptional co-activator binding protein, was up-regulated in the soleus muscle of high sucrose diet (HSD) induced insulin resistant rats and TNF-α exposed cultured myoblasts. Moreover, TNF-α induced phosphorylation of PIMT at the ERK1/2 target site Ser298. Wild type (WT) PIMT or phospho-mimic Ser298Asp mutant but not phospho-deficient Ser298Ala PIMT mutant abrogated insulin stimulated glucose uptake by L6 myotubes and neonatal rat skeletal myoblasts. Whereas, PIMT knock down relieved TNF-α inhibited insulin signaling. Mechanistic analysis revealed that PIMT differentially regulated the expression of GLUT4, MEF2A, PGC-1α and HDAC5 in cultured cells and skeletal muscle of Wistar rats. Further characterization showed that PIMT was recruited to GLUT4, MEF2A and HDAC5 promoters and overexpression of PIMT abolished the activity of WT but not MEF2A binding defective mutant GLUT4 promoter. Collectively, we conclude that PIMT mediates TNF-α induced insulin resistance at the skeletal muscle via the transcriptional modulation of GLUT4, MEF2A, PGC-1α and HDAC5 genes.
Collapse
|
19
|
Boon KL, Pearson MD, Koš M. Self-association of Trimethylguanosine Synthase Tgs1 is required for efficient snRNA/snoRNA trimethylation and pre-rRNA processing. Sci Rep 2015; 5:11282. [PMID: 26074133 PMCID: PMC4466884 DOI: 10.1038/srep11282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/21/2015] [Indexed: 12/23/2022] Open
Abstract
Trimethylguanosine Synthase catalyses transfer of two methyl groups to the m7G cap of RNA polymerase II transcribed snRNAs, snoRNAs, and telomerase RNA TLC1 to form a 2,2,7-trimethylguanosine cap. While in vitro studies indicate that Tgs1 functions as a monomer and the dimethylation of m7G caps is not a processive reaction, partially methylated sn(o)RNAs are typically not detected in living cells. Here we show that both yeast and human Tgs1p possess a conserved self-association property located at the N-terminus. A disruption of Tgs1 self-association led to a strong reduction of sn(o)RNA trimethylation as well as reduced nucleolar enrichment of Tgs1. Self-association of Tgs1p and its catalytic activity were also prerequisite to bypass the requirement for its accessory factor Swm2p for efficient pre-rRNA processing and snRNA trimethylation. The ability to self-associate might enable Tgs1 to efficiently dimethylate the caps of the targeted RNAs in vivo.
Collapse
Affiliation(s)
- Kum-Loong Boon
- 1] Biochemistry Center (BZH), University of Heidelberg, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany [2] Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Michael David Pearson
- Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Martin Koš
- Biochemistry Center (BZH), University of Heidelberg, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| |
Collapse
|
20
|
Siena LA, Ortiz JPA, Leblanc O, Pessino S. PnTgs1-like expression during reproductive development supports a role for RNA methyltransferases in the aposporous pathway. BMC PLANT BIOLOGY 2014; 14:297. [PMID: 25404464 PMCID: PMC4243328 DOI: 10.1186/s12870-014-0297-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 10/20/2014] [Indexed: 05/19/2023]
Abstract
BACKGROUND In flowering plants, apomixis (asexual reproduction via seeds) is widely believed to result from failure of key regulators of the sexual female reproductive pathway. In the past few years, both differential display and RNA-seq comparative approaches involving reproductive organs of sexual plants and their apomictic counterparts have yielded extensive lists of candidate genes. Nevertheless, only a limited number of these genes have been functionally characterized, with few clues consequently available for understanding the molecular control of apomixis. We have previously identified several cDNA fragments with high similarity to genes involved in RNA biology and with differential amplification between sexual and apomictic Paspalum notatum plants. Here, we report the characterization of one of these candidates, namely, N69 encoding a protein of the S-adenosyl-L-methionine-dependent methyltransferases superfamily. The purpose of this work was to extend the N69 cDNA sequence and to characterize its expression at different developmental stages in both sexual and apomictic individuals. RESULTS Molecular characterization of the N69 cDNA revealed homology with genes encoding proteins similar to yeast and mammalian trimethylguanosine synthase/PRIP-interacting proteins. These proteins play a dual role as ERK2-controlled transcriptional coactivators and mediators of sn(o)RNA and telomerase RNA cap trimethylation, and participate in mammals and yeast development. The N69-extended sequence was consequently renamed PnTgs1-like. Expression of PnTgs1-like during reproductive development was significantly higher in floral organs of sexual genotypes compared with apomicts. This difference was not detected in vegetative tissues. In addition, expression levels in reproductive tissues of several genotypes were negatively correlated with facultative apomixis rates. Moreover, in situ hybridization observations revealed that PnTgs1-like expression is relatively higher in ovules of sexual plants throughout development, from premeiosis to maturity. Tissues where differential expression is detected include nucellar cells, the site of aposporous initials differentiation in apomictic genotypes. CONCLUSIONS Our results indicate that PnTgs1-like (formerly N69) encodes a trimethylguanosine synthase-like protein whose function in mammals and yeast is critical for development, including reproduction. Our findings also suggest a pivotal role for this candidate gene in nucellar cell fate, as its diminished expression is correlated with initiation of the apomictic pathway in plants.
Collapse
Affiliation(s)
- Lorena A Siena
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
| | - Juan Pablo A Ortiz
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
- />Instituto de Botánica del Nordeste -IBONE- (UNNE-CONICET), Facultad de Ciencias Agrarias, Universidad Nacional del Nordeste, Sargento Cabral 2131, 3400 Corrientes, Argentina
| | - Olivier Leblanc
- />Institut de Recherche pour le Développement, ERL 5300 IRD/CNRS, UMR 232 IRD/Université de Montpellier 2, 911 Avenue Agropolis, Montpellier, France
| | - Silvina Pessino
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
| |
Collapse
|
21
|
Ohlmann T, Mengardi C, López-Lastra M. Translation initiation of the HIV-1 mRNA. ACTA ACUST UNITED AC 2014; 2:e960242. [PMID: 26779410 DOI: 10.4161/2169074x.2014.960242] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/23/2014] [Accepted: 06/17/2014] [Indexed: 12/17/2022]
Abstract
Translation initiation of the full-length mRNA of the human immunodeficiency virus can occur via several different mechanisms to maintain production of viral structural proteins throughout the replication cycle. HIV-1 viral protein synthesis can occur by the use of both a cap-dependant and IRES-driven mechanism depending on the physiological conditions of the cell and the status of the ongoing infection. For both of these mechanisms there is a need for several viral and cellular co-factors for optimal translation of the viral mRNA. In this review we will describe the mechanism used by the full-length mRNA to initiate translation highlighting the role of co-factors within this process. A particular emphasis will be given to the role of the DDX3 RNA helicase in HIV-1 mRNA translation initiation.
Collapse
Affiliation(s)
- Théophile Ohlmann
- CIRI; International Center for Infectiology Research; Université de Lyon; Lyon, France; Inserm; Lyon, France; Ecole Normale Supérieure de Lyon; Lyon, France; Université Lyon 1; Center International de Recherche en Infectiologie; Lyon, France; CNRS; Lyon, France
| | - Chloé Mengardi
- CIRI; International Center for Infectiology Research; Université de Lyon; Lyon, France; Inserm; Lyon, France; Ecole Normale Supérieure de Lyon; Lyon, France; Université Lyon 1; Center International de Recherche en Infectiologie; Lyon, France; CNRS; Lyon, France
| | - Marcelo López-Lastra
- Laboratorio de Virología Molecular; Instituto Milenio de Inmunología e Inmunoterapia; Centro de Investigaciones Médicas; Escuela de Medicina; Pontificia Universidad Católica de Chile ; Santiago, Chile
| |
Collapse
|
22
|
Schulz D, Rentmeister A. An enzyme-coupled high-throughput assay for screening RNA methyltransferase activity inE. Colicell lysate. RNA Biol 2014; 9:577-86. [DOI: 10.4161/rna.19818] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
23
|
Kapadia B, Viswakarma N, Parsa KVL, Kain V, Behera S, Suraj SK, Babu PP, Kar A, Panda S, Zhu YJ, Jia Y, Thimmapaya B, Reddy JK, Misra P. ERK2-mediated phosphorylation of transcriptional coactivator binding protein PIMT/NCoA6IP at Ser298 augments hepatic gluconeogenesis. PLoS One 2013; 8:e83787. [PMID: 24358311 PMCID: PMC3866170 DOI: 10.1371/journal.pone.0083787] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/08/2013] [Indexed: 12/22/2022] Open
Abstract
PRIP-Interacting protein with methyl transferase domain (PIMT) serves as a molecular bridge between CREB-binding protein (CBP)/ E1A binding protein p300 (Ep300) -anchored histone acetyl transferase and the Mediator complex sub-unit1 (Med1) and modulates nuclear receptor transcription. Here, we report that ERK2 phosphorylates PIMT at Ser(298) and enhances its ability to activate PEPCK promoter. We observed that PIMT is recruited to PEPCK promoter and adenoviral-mediated over-expression of PIMT in rat primary hepatocytes up-regulated expression of gluconeogenic genes including PEPCK. Reporter experiments with phosphomimetic PIMT mutant (PIMT(S298D)) suggested that conformational change may play an important role in PIMT-dependent PEPCK promoter activity. Overexpression of PIMT and Med1 together augmented hepatic glucose output in an additive manner. Importantly, expression of gluconeogenic genes and hepatic glucose output were suppressed in isolated liver specific PIMT knockout mouse hepatocytes. Furthermore, consistent with reporter experiments, PIMT(S298D) but not PIMT(S298A) augmented hepatic glucose output via up-regulating the expression of gluconeogenic genes. Pharmacological blockade of MAPK/ERK pathway using U0126, abolished PIMT/Med1-dependent gluconeogenic program leading to reduced hepatic glucose output. Further, systemic administration of T4 hormone to rats activated ERK1/2 resulting in enhanced PIMT ser(298) phosphorylation. Phosphorylation of PIMT led to its increased binding to the PEPCK promoter, increased PEPCK expression and induction of gluconeogenesis in liver. Thus, ERK2-mediated phosphorylation of PIMT at Ser(298) is essential in hepatic gluconeogenesis, demonstrating an important role of PIMT in the pathogenesis of hyperglycemia.
Collapse
Affiliation(s)
- Bandish Kapadia
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Navin Viswakarma
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Kishore V. L. Parsa
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Vasundhara Kain
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Soma Behera
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Sashidhara Kaimal Suraj
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Phanithi Prakash Babu
- Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, Andhra Pradesh, India
| | - Anand Kar
- Department of Life Sciences, Devi Ahilya University, Indore, Madhya Pradesh, India
| | - Sunanda Panda
- Department of Life Sciences, Devi Ahilya University, Indore, Madhya Pradesh, India
| | - Yi-jun Zhu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Janardan K. Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- * E-mail: (PM); (JKR)
| | - Parimal Misra
- Department of Biology, Dr Reddy’s Institute of Life Sciences, An Associate Institute of University of Hyderabad, Hyderabad, Andhra Pradesh, India
- * E-mail: (PM); (JKR)
| |
Collapse
|
24
|
Zhu Y, Feng F, Yu J, Song B, Hu M, Gao X, Wang Y, Zhang Q. L1-ORF1p, a Smad4 interaction protein, promotes proliferation of HepG2 cells and tumorigenesis in mice. DNA Cell Biol 2013; 32:531-40. [PMID: 23863096 DOI: 10.1089/dna.2013.2097] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Long interspersed nucleotide element (LINE-1; L1) as an autonomous retrotransposon is localized usually in AT-rich, low-recombined, and gene-poor regions of genome. It is transiently activated in embryonic development and continuously activated in all tumor cells tested so far. Full-length L1 gene contains 5' untranslated region, two open reading frames (ORFs) encoded L1ORF1p and L1ORF2p, and a 3' terminal polyadenylation site. Compared with L1ORF2p, a protein encompassing reverse transcriptase and endonuclease activities, L1ORF1p remains to be elucidated. With liver cancer cells and tissues, the expression and sub-localization of L1ORF1p were investigated and shown that L1-ORF1p expresses just in liver cancer cells and tissues but not in normal liver cells and almost not in adjacent tissues. To characterize L1ORF1p, the strategies for over-expression and down-regulation of L1ORF1p in transfected cells were implemented. The phenomenon of promoting cell proliferation and colony formation was observed in transfected cells with L1ORF1p over-expression and vice versa. Down-regulation of L1ORF1p suppresses tumorigenesis in vitro and in vivo. Smad4 as an interaction protein of L1ORF1p is identified for the first time, while L1ORF1p is responsible for Smad4 sequestration in the cytoplasm. Thus, L1ORF1p contributed to tumorigenesis and may attribute to, at least partly, its participation in Smad4-signaling regulation.
Collapse
Affiliation(s)
- Yunfeng Zhu
- The Institute of Life Sciences and Bio-Engineering in Beijing Jiaotong University, Beijing, China.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Misra P, Viswakarma N, Reddy JK. Peroxisome proliferator-activated receptor-α signaling in hepatocarcinogenesis. Subcell Biochem 2013; 69:77-99. [PMID: 23821144 DOI: 10.1007/978-94-007-6889-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Peroxisomes are subcellular organelles that are found in the cytoplasm of most animal cells. They perform diverse metabolic functions, including H2O2-derived respiration, β-oxidation of fatty acids, and cholesterol metabolism. Peroxisome proliferators are a large class of structurally dissimilar industrial and pharmaceutical chemicals that were originally identified as inducers of both the size and the number of peroxisomes in rat and mouse livers or hepatocytes in vitro. Exposure to peroxisome proliferators leads to a stereotypical orchestration of adaptations consisting of hepatocellular hypertrophy and hyperplasia, and transcriptional induction of fatty acid metabolizing enzymes regulated in parallel with peroxisome proliferation. Chronic exposure to peroxisome proliferators causes liver tumors in both male and female mice and rats. Evidence indicates a pivotal role for a subset of nuclear receptor superfamily members, called peroxisome proliferator-activated receptors (PPARs), in mediating energy metabolism. Upon activation, PPARs regulate the expression of genes involved in lipid metabolism and peroxisome proliferation, as well as genes involved in cell growth. In this review, we describe the molecular mode of action of PPAR transcription factors, including ligand binding, interaction with specific DNA response elements, transcriptional activation, and cross talk with other signaling pathways. We discuss the evidence that suggests that PPARα and transcriptional coactivator Med1/PBP, a key subunit of the Mediator complex play a central role in mediating hepatic steatosis to hepatocarcinogenesis. Disproportionate increases in H2O2-generating enzymes generates excess reactive oxygen species resulting in sustained oxidative stress and progressive endoplasmic reticulum (ER) stress with activation of unfolded protein response signaling. Thus, these major contributors coupled with hepatocellular proliferation are the key players of peroxisome proliferators-induced hepatocarcinogenesis.
Collapse
Affiliation(s)
- Parimal Misra
- Department of Biology, Dr. Reddy's Institute of Life Sciences, An Associate Institute of University of Hyderabad, Gachibowli, Hyderabad, 500046, India,
| | | | | |
Collapse
|
26
|
Jia Y, Viswakarma N, Crawford SE, Sarkar J, Sambasiva Rao M, Karpus WJ, Kanwar YS, Zhu YJ, Reddy JK. Early embryonic lethality of mice with disrupted transcription cofactor PIMT/NCOA6IP/Tgs1 gene. Mech Dev 2012; 129:193-207. [PMID: 22982455 DOI: 10.1016/j.mod.2012.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 08/09/2012] [Accepted: 08/27/2012] [Indexed: 11/29/2022]
Abstract
PIMT (also known as PIPMT/NCOA6IP/Tgs1), first isolated as a transcription coactivator PRIP (NCOA6)-interacting 96-kDa protein with RNA-binding property, possesses RNA methyltransferase activity. As a transcription coactivator binding protein, PIMT enhances the nuclear receptor transcriptional activity and its methyltransferase property is involved in the formation of the 2,2,7-trimethylguanosine cap of non-coding small RNAs, but the in vivo functions of this gene have not been fully explored. To elucidate the biological functions, we used gene targeting to generate mice with a disrupted PIMT/Tgs1 gene. Disruption of PIMT gene results in early embryonic lethality due to impairment of development around the blastocyst and uterine implantation stages. We show that PIMT is expressed in all cells of the E3.5day blastocyst in the mouse. PIMT null mutation abolished PIMT expression in all cells of the blastocyst and caused a reduction in the expression of Oct4 and Nanog transcription factor proteins in the E3.5 blastocyst resulting in the near failure to form inner cell mass (ICM). With conditional deletion of PIMT gene, mouse embryonic fibroblasts (MEFs) exhibit defective wound healing in the scratch assay and a reduction in cell proliferation due to decreased G₀/G₁ transition and G₂/M phase cell cycle arrest. We conclude that PIMT/NCOA6IP, which is expressed in all cells of the 3.5 day stage blastocyst, is indispensable for early embryonic development.
Collapse
Affiliation(s)
- Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Angénieux C, Waharte F, Gidon A, Signorino-Gelo F, Wurtz V, Hojeij R, Proamer F, Gachet C, Van Dorsselaer A, Hanau D, Salamero J, de la Salle H. Lysosomal-associated transmembrane protein 5 (LAPTM5) is a molecular partner of CD1e. PLoS One 2012; 7:e42634. [PMID: 22880058 PMCID: PMC3411835 DOI: 10.1371/journal.pone.0042634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/10/2012] [Indexed: 11/24/2022] Open
Abstract
The CD1e protein participates in the presentation of lipid antigens in dendritic cells. Its transmembrane precursor is transported to lysosomes where it is cleaved into an active soluble form. In the presence of bafilomycin, which inhibits vacuolar ATPase and consequently the acidification of endosomal compartments, CD1e associates with a 27 kD protein. In this work, we identified this molecular partner as LAPTM5. The latter protein and CD1e colocalize in trans-Golgi and late endosomal compartments. The quantity of LAPTM5/CD1e complexes increases when the cells are treated with bafilomycin, probably due to the protection of LAPTM5 from lysosomal proteases. Moreover, we could demonstrate that LAPTM5/CD1e association occurs under physiological conditions. Although LAPTM5 was previously shown to act as a platform recruiting ubiquitin ligases and facilitating the transport of receptors to lysosomes, we found no evidence that LATPM5 controls either CD1e ubiquitination or the generation of soluble lysosomal CD1e proteins. Notwithstanding these last observations, the interaction of LAPTM5 with CD1e and their colocalization in antigen processing compartments both suggest that LAPTM5 might influence the role of CD1e in the presentation of lipid antigens.
Collapse
Affiliation(s)
- Catherine Angénieux
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - François Waharte
- Cell and Tissue Imaging Facility, Unité Mixte de Recherche 144, CNRS-Institut Curie Section de Recherche, Paris, France
| | - Alexandre Gidon
- Molecular mechanisms of intracellular transport, Unité Mixte de Recherche 144, CNRS-Institut Curie Section de Recherche, Paris, France
| | - François Signorino-Gelo
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Virginie Wurtz
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S949, Strasbourg, France
| | - Rim Hojeij
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Fabienne Proamer
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Christian Gachet
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S949, Strasbourg, France
| | - Alain Van Dorsselaer
- Université de Strasbourg, Strasbourg, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7178, Strasbourg F-67037, France
| | - Daniel Hanau
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Jean Salamero
- Cell and Tissue Imaging Facility, Unité Mixte de Recherche 144, CNRS-Institut Curie Section de Recherche, Paris, France
- Molecular mechanisms of intracellular transport, Unité Mixte de Recherche 144, CNRS-Institut Curie Section de Recherche, Paris, France
| | - Henri de la Salle
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S725, Strasbourg, France
- Etablissement Français du Sang-Alsace, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
28
|
Chemical approaches for structure and function of RNA in postgenomic era. J Nucleic Acids 2012; 2012:369058. [PMID: 22347623 PMCID: PMC3278928 DOI: 10.1155/2012/369058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 06/21/2011] [Accepted: 07/05/2011] [Indexed: 01/11/2023] Open
Abstract
In the study of cellular RNA chemistry, a major thrust of research focused upon sequence determinations for decades. Structures of snRNAs (4.5S RNA I (Alu), U1, U2, U3, U4, U5, and U6) were determined at Baylor College of Medicine, Houston, Tex, in an earlier time of pregenomic era. They show novel modifications including base methylation, sugar methylation, 5′-cap structures (types 0–III) and sequence heterogeneity. This work offered an exciting problem of posttranscriptional modification and underwent numerous significant advances through technological revolutions during pregenomic, genomic, and postgenomic eras. Presently, snRNA research is making progresses involved in enzymology of snRNA modifications, molecular evolution, mechanism of spliceosome assembly, chemical mechanism of intron removal, high-order structure of snRNA in spliceosome, and pathology of splicing. These works are destined to reach final pathway of work “Function and Structure of Spliceosome” in addition to exciting new exploitation of other noncoding RNAs in all aspects of regulatory functions.
Collapse
|
29
|
Powell E, Kuhn P, Xu W. Nuclear Receptor Cofactors in PPARgamma-Mediated Adipogenesis and Adipocyte Energy Metabolism. PPAR Res 2011; 2007:53843. [PMID: 17389765 PMCID: PMC1783724 DOI: 10.1155/2007/53843] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 10/17/2006] [Accepted: 10/17/2006] [Indexed: 01/18/2023] Open
Abstract
Transcriptional cofactors are integral to the proper function and regulation of nuclear receptors. Members of the peroxisome proliferator-activated receptor (PPAR) family of nuclear receptors are involved in the regulation of lipid and carbohydrate metabolism. They modulate gene transcription in response to a wide variety of ligands, a process that is mediated by transcriptional coactivators and corepressors. The mechanisms by which these cofactors mediate transcriptional regulation of nuclear receptor function are still being elucidated. The rapidly increasing array of cofactors has brought into focus the need for a clear understanding of how these cofactors interact in ligand- and cell-specific manners. This review highlights the differential effects of the assorted cofactors regulating the transcriptional action of PPARγ and summarizes the recent advances in understanding the physiological functions of corepressors and coactivators.
Collapse
Affiliation(s)
- Emily Powell
- McArdle Laboratory for Cancer Research, University of Wisconsin, 1400 University Avenue, Madison, WI 53706, USA
| | - Peter Kuhn
- McArdle Laboratory for Cancer Research, University of Wisconsin, 1400 University Avenue, Madison, WI 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin, 1400 University Avenue, Madison, WI 53706, USA
- *Wei Xu:
| |
Collapse
|
30
|
PRIC295, a Nuclear Receptor Coactivator, Identified from PPARα-Interacting Cofactor Complex. PPAR Res 2010; 2010. [PMID: 20885938 PMCID: PMC2946606 DOI: 10.1155/2010/173907] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 07/22/2010] [Indexed: 01/22/2023] Open
Abstract
The peroxisome proliferator-activated receptor-α (PPARα) plays a key role in lipid metabolism and energy combustion. Chronic activation of PPARα in rodents leads to the development of hepatocellular carcinomas. The ability of PPARα to induce expression of its target genes depends on Mediator, an evolutionarily conserved complex of cofactors and, in particular, the subunit 1 (Med1) of this complex. Here, we report the identification and characterization of PPARα-interacting cofactor (PRIC)-295 (PRIC295), a novel coactivator protein, and show that it interacts with the Med1 and Med24 subunits of the Mediator complex. PRIC295 contains 10 LXXLL signature motifs that facilitate nuclear receptor binding and interacts with PPARα and five other members of the nuclear receptor superfamily in a ligand-dependent manner. PRIC295 enhances the transactivation function of PPARα, PPARγ, and ERα. These data demonstrate that PRIC295 interacts with nuclear receptors such as PPARα and functions as a transcription coactivator under in vitro conditions and may play an important role in mediating the effects in vivo as a member of the PRIC complex with Med1 and Med24.
Collapse
|
31
|
Molecular Mechanisms and Genome-Wide Aspects of PPAR Subtype Specific Transactivation. PPAR Res 2010; 2010. [PMID: 20862367 PMCID: PMC2938449 DOI: 10.1155/2010/169506] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 06/27/2010] [Indexed: 12/13/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are central regulators of fat metabolism, energy homeostasis, proliferation, and inflammation. The three PPAR subtypes, PPARα, β/δ, and γ activate overlapping but also very different target gene programs. This review summarizes the insights into PPAR subtype-specific transactivation provided by genome-wide studies and discusses the recent advances in the understanding of the molecular mechanisms underlying PPAR subtype specificity with special focus on the regulatory role of AF-1.
Collapse
|
32
|
Abstract
Peroxisome proliferator-activated receptor (PPAR)alpha, beta (also known as delta), and gamma function as sensors for fatty acids and fatty acid derivatives and control important metabolic pathways involved in the maintenance of energy balance. PPARs also regulate other diverse biological processes such as development, differentiation, inflammation, and neoplasia. In the nucleus, PPARs exist as heterodimers with retinoid X receptor-alpha bound to DNA with corepressor molecules. Upon ligand activation, PPARs undergo conformational changes that facilitate the dissociation of corepressor molecules and invoke a spatiotemporally orchestrated recruitment of transcription cofactors including coactivators and coactivator-associated proteins. While a given nuclear receptor regulates the expression of a prescribed set of target genes, coactivators are likely to influence the functioning of many regulators and thus affect the transcription of many genes. Evidence suggests that some of the coactivators such as PPAR-binding protein (PBP/PPARBP), thyroid hormone receptor-associated protein 220 (TRAP220), and mediator complex subunit 1 (MED1) may exert a broader influence on the functions of several nuclear receptors and their target genes. Investigations into the role of coactivators in the function of PPARs should strengthen our understanding of the complexities of metabolic diseases associated with energy metabolism.
Collapse
|
33
|
Trimethylguanosine capping selectively promotes expression of Rev-dependent HIV-1 RNAs. Proc Natl Acad Sci U S A 2010; 107:14787-92. [PMID: 20679221 DOI: 10.1073/pnas.1009490107] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
5'-mRNA capping is an early modification that affects pre-mRNA synthesis/splicing, RNA cytoplasmic transport, and mRNA translation and turnover. In eukaryotes, a 7-methylguanosine (m7G) cap is added to newly transcribed RNA polymerase II (RNAP II) transcripts. A subset of RNAP II-transcribed cellular RNAs, including small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), and telomerase RNA, is further hypermethylated at the exocyclic N2 of the guanosine to create a trimethylguanosine (TMG)-capped RNA. Some of these TMG-capped RNAs are transported within the nucleus and from the nucleus to the cytoplasm by the CRM-1 (required for chromosome region maintenance) protein. CRM-1 is also used to export Rev/RRE-dependent unspliced/ partially spliced HIV-1 RNAs. Here we report that like snRNAs and snoRNAs, some Rev/RRE-dependent HIV-1 RNAs are TMG-capped. The methyltransferase responsible for TMG modification of HIV-1 RNAs is the human PIMT (peroxisome proliferator-activated receptor-interacting protein with methyltransferase) protein. TMG capping of unspliced/partially spliced HIV-1 RNAs represents a new regulatory mechanism for selective expression.
Collapse
|
34
|
Singh AK, Battu A, Mohareer K, Hasnain SE, Ehtesham NZ. Transcription of human resistin gene involves an interaction of Sp1 with peroxisome proliferator-activating receptor gamma (PPARgamma). PLoS One 2010; 5:e9912. [PMID: 20360975 PMCID: PMC2848011 DOI: 10.1371/journal.pone.0009912] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 02/26/2010] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Resistin is a cysteine rich protein, mainly expressed and secreted by circulating human mononuclear cells. While several factors responsible for transcription of mouse resistin gene have been identified, not much is known about the factors responsible for the differential expression of human resistin. METHODOLOGY/PRINCIPAL FINDING We show that the minimal promoter of human resistin lies within approximately 80 bp sequence upstream of the transcriptional start site (-240) whereas binding sites for cRel, CCAAT enhancer binding protein alpha (C/EBP-alpha), activating transcription factor 2 (ATF-2) and activator protein 1 (AP-1) transcription factors, important for induced expression, are present within sequences up to -619. Specificity Protein 1(Sp1) binding site (-276 to -295) is also present and an interaction of Sp1 with peroxisome proliferator activating receptor gamma (PPARgamma) is necessary for constitutive expression in U937 cells. Indeed co-immunoprecipitation assay demonstrated a direct physical interaction of Sp1 with PPARgamma in whole cell extracts of U937 cells. Phorbol myristate acetate (PMA) upregulated the expression of resistin mRNA in U937 cells by increasing the recruitment of Sp1, ATF-2 and PPARgamma on the resistin gene promoter. Furthermore, PMA stimulation of U937 cells resulted in the disruption of Sp1 and PPARgamma interaction. Chromatin immunoprecipitation (ChIP) assay confirmed the recruitment of transcription factors phospho ATF-2, Sp1, Sp3, PPARgamma, chromatin modifier histone deacetylase 1 (HDAC1) and the acetylated form of histone H3 but not cRel, C/EBP-alpha and phospho c-Jun during resistin gene transcription. CONCLUSION Our findings suggest a complex interplay of Sp1 and PPARgamma along with other transcription factors that drives the expression of resistin in human monocytic U937 cells.
Collapse
Affiliation(s)
| | - Aruna Battu
- National Institute of Nutrition, Indian Council for Medical Research, Hyderabad, India
| | | | - Seyed E. Hasnain
- University of Hyderabad, Hyderabad, India
- Institute of Life Sciences, University of Hyderabad, Hyderabad, India
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Nasreen Z. Ehtesham
- National Institute of Nutrition, Indian Council for Medical Research, Hyderabad, India
- Institute of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
35
|
Powell CL, Bradford BU, Craig CP, Tsuchiya M, Uehara T, O'Connell TM, Pogribny IP, Melnyk S, Koop DR, Bleyle L, Threadgill DW, Rusyn I. Mechanism for prevention of alcohol-induced liver injury by dietary methyl donors. Toxicol Sci 2010; 115:131-9. [PMID: 20118189 DOI: 10.1093/toxsci/kfq031] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alcohol-induced liver injury (ALI) has been associated with, among other molecular changes, abnormal hepatic methionine metabolism, resulting in decreased levels of S-adenosylmethionine (SAM). Dietary methyl donor supplements such as SAM and betaine mitigate ALI in animal models; however, the mechanisms of protection remain elusive. It has been suggested that methyl donors may act via attenuation of alcohol-induced oxidative stress. We hypothesized that the protective action of methyl donors is mediated by an effect on the oxidative metabolism of alcohol in the liver. Male C57BL/6J mice were administered a control high-fat diet or diet enriched in methyl donors with or without alcohol for 4 weeks using the enteral alcohol feeding model. As expected, attenuation of ALI and an increase in reduced glutathione:oxidized glutathione ratio were achieved with methyl donor supplementation. Interestingly, methyl donors led to a 35% increase in blood alcohol elimination rate, and while there was no effect on alcohol metabolism in the stomach, a profound effect on liver alcohol metabolism was observed. The catalase-dependent pathway of alcohol metabolism was induced, yet the increase in CYP2E1 activity by alcohol was blunted, which may be mitigating production of oxidants. Additional factors contributing to the protective effects of methyl donors in ALI were increased activity of low- and high-K(m) aldehyde dehydrogenases leading to lower hepatic acetaldehyde, maintenance of the efficient mitochondrial energy metabolism, and promotion of peroxisomal beta-oxidation. Profound changes in alcohol metabolism represent additional important mechanism of the protective effect of methyl donors in ALI.
Collapse
Affiliation(s)
- Christine L Powell
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Roles of histone H3-lysine 4 methyltransferase complexes in NR-mediated gene transcription. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:343-82. [PMID: 20374709 DOI: 10.1016/s1877-1173(09)87010-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Transcriptional regulation by nuclear hormone receptors (NRs) requires multiple coregulators that modulate chromatin structures by catalyzing a diverse array of posttranslational modifications of histones. Different combinations of these modifications yield dynamic functional outcomes, constituting an epigenetic histone code. This code is inscribed by histone-modifying enzymes and decoded by effector proteins that recognize specific covalent marks. One important modification associated with active chromatin structures is methylation of histone H3-lysine 4 (H3K4). Crucial roles for this modification in NR transactivation have been recently highlighted through our purification and subsequent characterization of a steady-state complex associated with ASC-2, a coactivator of NRs and other transcription factors. This complex, designated ASCOM for ASC-2 complex, contains H3K4-methyltransferase MLL3/HALR or its paralogue MLL4/ALR and represents the first Set1-like H3K4-methyltransferase complex to be reported in vertebrates. This review focuses on recent progress in our understanding of how ASCOM-MLL3 and ASCOM-MLL4 influence NR-mediated gene transcription and of their physiological function.
Collapse
|
37
|
Colley SM, Leedman PJ. SRA and its binding partners: an expanding role for RNA-binding coregulators in nuclear receptor-mediated gene regulation. Crit Rev Biochem Mol Biol 2009; 44:25-33. [PMID: 19280430 DOI: 10.1080/10409230802661719] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The discovery that SRA RNA can function as a nuclear receptor (NR) coactivator resulted in a fundamental change in the perception of how NRs and their coregulators may regulate hormone signaling pathways. The subsequent identification of molecules capable of binding SRA, including SHARP, p68, and more recently SLIRP, which also function as coregulators, has further broadened our understanding of NR-dependent gene regulation. The integral role that NRs play in directing developmental, metabolic and pathological programs of transcription has defined them as paramount targets for treating a broad range of human diseases. Thus with a greater understanding of SRA and its interactions with its binding partners, novel RNA-protein interactions may be identified and exploited for therapeutic gain. Here we discuss the isolation of SRA, its impact on NR activity and interactions with known binding partners.
Collapse
Affiliation(s)
- Shane M Colley
- University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | | |
Collapse
|
38
|
Monecke T, Dickmanns A, Ficner R. Structural basis for m7G-cap hypermethylation of small nuclear, small nucleolar and telomerase RNA by the dimethyltransferase TGS1. Nucleic Acids Res 2009; 37:3865-77. [PMID: 19386620 PMCID: PMC2709555 DOI: 10.1093/nar/gkp249] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The 5′-cap of spliceosomal small nuclear RNAs, some small nucleolar RNAs and of telomerase RNA was found to be hypermethylated in vivo. The Trimethylguanosine Synthase 1 (TGS1) mediates this conversion of the 7-methylguanosine-cap to the 2,2,7-trimethylguanosine (m3G)-cap during maturation of the RNPs. For mammalian UsnRNAs the generated m2,2,7G-cap is one part of a bipartite import signal mediating the transport of the UsnRNP-core complex into the nucleus. In order to understand the structural organization of human TGS1 as well as substrate binding and recognition we solved the crystal structure of the active TGS1 methyltransferase domain containing both, the minimal substrate m7GTP and the reaction product S-adenosyl-l-homocysteine (AdoHcy). The methyltransferase of human TGS1 harbors the canonical class 1 methyltransferase fold as well as an unique N-terminal, α-helical domain of 40 amino acids, which is essential for m7G-cap binding and catalysis. The crystal structure of the substrate bound methyltransferase domain as well as mutagenesis studies provide insight into the catalytic mechanism of TGS1.
Collapse
Affiliation(s)
- Thomas Monecke
- Abteilung für Molekulare Strukturbiologie, Institut für Mikrobiologie und Genetik, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| | | | | |
Collapse
|
39
|
Zhu YT, Hu L, Qi C, Zhu YJ. PRIP promotes tumor formation through enhancing serum-responsive factor-mediated FOS expression. J Biol Chem 2009; 284:14485-92. [PMID: 19329434 DOI: 10.1074/jbc.m900935200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
PRIP (peroxisome proliferator-activator receptor interacting protein) is a nuclear receptor coactivator required for mammary gland development. To understand the function of PRIP in breast tumorigenesis, we established a mammary tumor cell line with the PRIP(Loxp/Loxp) genotype. By knocking out the PRIP gene in the tumor cell line, we demonstrated that PRIP deficiency led to inhibited tumor formation without affecting tumor cell proliferation. The PRIP deficiency was associated with decreased cell invasion and migration capabilities. We found that PRIP deficiency substantially reduced FOS gene expression. A chromatin immunoprecipitation assay revealed that PRIP was recruited to the FOS promoter. In addition, we demonstrated that PRIP also directly up-regulated the FOS gene expression in human breast cancer cells. Promoter analysis showed that PRIP acted through serum-responsive factor to regulate FOS gene expression. Finally, by re-expressing the FOS gene, we confirmed that the inhibited tumor formation of PRIP-deficient tumor cells was due to reduced expression of the FOS gene.
Collapse
Affiliation(s)
- Yiwei Tony Zhu
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
40
|
In vitro characterization of the enzyme properties of the phospholipid N-methyltransferase PmtA from Agrobacterium tumefaciens. J Bacteriol 2009; 191:2033-41. [PMID: 19181804 DOI: 10.1128/jb.01591-08] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Agrobacterium tumefaciens requires phosphatidylcholine (PC) in its membranes for plant infection. The phospholipid N-methyltransferase PmtA catalyzes all three transmethylation reactions of phosphatidylethanolamine (PE) to PC via the intermediates monomethylphosphatidylethanolamine (MMPE) and dimethylphosphatidylethanolamine (DMPE). The enzyme uses S-adenosylmethionine (SAM) as the methyl donor, converting it to S-adenosylhomocysteine (SAH). Little is known about the activity of bacterial Pmt enzymes, since PC biosynthesis in prokaryotes is rare. In this article, we present the purification and in vitro characterization of A. tumefaciens PmtA, which is a monomeric protein. It binds to PE, the intermediates MMPE and DMPE, the end product PC, and phosphatidylglycerol (PG) and phosphatidylinositol. Binding of the phospholipid substrates precedes binding of SAM. We used a coupled in vitro assay system to demonstrate the enzymatic activity of PmtA and to show that PmtA is inhibited by the end products PC and SAH and the antibiotic sinefungin. The presence of PG stimulates PmtA activity. Our study provides insights into the catalysis and control of a bacterial phospholipid N-methyltransferase.
Collapse
|
41
|
Hausmann S, Zheng S, Costanzo M, Brost RL, Garcin D, Boone C, Shuman S, Schwer B. Genetic and biochemical analysis of yeast and human cap trimethylguanosine synthase: functional overlap of 2,2,7-trimethylguanosine caps, small nuclear ribonucleoprotein components, pre-mRNA splicing factors, and RNA decay pathways. J Biol Chem 2008; 283:31706-18. [PMID: 18775984 PMCID: PMC2581544 DOI: 10.1074/jbc.m806127200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Revised: 09/04/2008] [Indexed: 12/26/2022] Open
Abstract
Trimethylguanosine synthase (Tgs1) is the enzyme that converts standard m(7)G caps to the 2,2,7-trimethylguanosine (TMG) caps characteristic of spliceosomal small nuclear RNAs. Fungi and mammalian somatic cells are able to grow in the absence of Tgs1 and TMG caps, suggesting that an essential function of the TMG cap might be obscured by functional redundancy. A systematic screen in budding yeast identified nonessential genes that, when deleted, caused synthetic growth defects with tgs1Delta. The Tgs1 interaction network embraced proteins implicated in small nuclear ribonucleoprotein function and spliceosome assembly, including Mud2, Nam8, Brr1, Lea1, Ist3, Isy1, Cwc21, and Bud13. Complementation of the synthetic lethality of mud2Delta tgs1Delta and nam8Delta tgs1Delta strains by wild-type TGS1, but not by catalytically defective mutants, indicated that the TMG cap is essential for mitotic growth when redundant splicing factors are missing. Our genetic analysis also highlighted synthetic interactions of Tgs1 with proteins implicated in RNA end processing and decay (Pat1, Lsm1, and Trf4) and regulation of polymerase II transcription (Rpn4, Spt3, Srb2, Soh1, Swr1, and Htz1). We find that the C-terminal domain of human Tgs1 can function in lieu of the yeast protein in vivo. We present a biochemical characterization of the human Tgs1 guanine-N2 methyltransferase reaction and identify individual amino acids required for methyltransferase activity in vitro and in vivo.
Collapse
Affiliation(s)
- Stéphane Hausmann
- Department of Microbiology and Molecular Medicine, University of Geneva, CH1211 Geneva, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Franke J, Gehlen J, Ehrenhofer-Murray AE. Hypermethylation of yeast telomerase RNA by the snRNA and snoRNA methyltransferase Tgs1. J Cell Sci 2008; 121:3553-60. [PMID: 18840651 DOI: 10.1242/jcs.033308] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Telomerase in Saccharomyces cerevisiae consists of three protein subunits and the RNA moiety TLC1, which together ensure the complete replication of chromosome ends. TLC1 shares several features with snRNA, among them the presence of a trimethylguanosine (m(3)G) cap structure at the 5' end of the RNA. Here, we report that the yeast snRNA and snoRNA methyltransferase Tgs1 is responsible for TLC1 m(3)G cap formation. The absence of Tgs1 caused changes in telomere length and structure, improved telomeric silencing and stabilized telomeric recombination. Genetic analyses implicated a role for the TLC1 m(3)G cap in the coordination between telomerase and DNA polymerase for end replication. Furthermore, tgs1Delta cells displayed a shortened replicative lifespan, suggesting that the loss of the m(3)G cap of TLC1 causes premature aging.
Collapse
Affiliation(s)
- Jacqueline Franke
- Zentrum für Medizinische Biotechnologie, Universität Duisburg-Essen, Universitätsstr. 5, 45117 Essen, Germany
| | | | | |
Collapse
|
43
|
Surapureddi S, Rana R, Reddy JK, Goldstein JA. Nuclear receptor coactivator 6 mediates the synergistic activation of human cytochrome P-450 2C9 by the constitutive androstane receptor and hepatic nuclear factor-4alpha. Mol Pharmacol 2008; 74:913-23. [PMID: 18552123 PMCID: PMC3815599 DOI: 10.1124/mol.108.048983] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nuclear receptor coactivator 6 (NCOA6) also known as PRIP/RAP250/ASC-2 anchors a steady-state complex of cofactors and function as a transcriptional coactivator for certain nuclear receptors. This is the first study to identify NCOA6 as a hepatic nuclear factor 4alpha (HNF4alpha)-interacting protein. CYP2C9 is an important enzyme that metabolizes both commonly used therapeutic drugs and important endogenous compounds. We have shown previously that constitutive androstane receptor (CAR) (a xenobiotic-sensing receptor) up-regulates the CYP2C9 promoter through binding to a distal site, whereas HNF4alpha transcriptionally up-regulates CYP2C9 via proximal sites. We demonstrate ligand-enhanced synergistic cross-talk between CAR and HNF4alpha. We identify NCOA6 as crucial to the underlying mechanism of this cross-talk. NCOA6 was identified as an HNF4alpha-interacting protein in this study using a yeast two-hybrid screen and GST pull-down assays. Furthermore, we identified NCOA6, CAR, and other coactivators as part of a mega complex of cofactors associated with HNF4alpha in HepG2 cells. Although the interaction of NCOA6 with CAR is specifically through the first LXXLL motif of NCOA6, both LXXLL motifs are involved in its interaction with HNF4alpha. Silencing of NCOA6 abrogated the synergistic activation of the CYP2C9 promoter and the synergistic induction of the CYP2C9 gene by CAR-HNF4alpha. Chromatin immunoprecipitation analysis revealed that NCOA6 can pull down both the proximal HNF4alpha and distal CAR binding sites of the CYP2C9 promoter and provides the basis for the recruitment of other cofactors. We conclude that the coactivator NCOA6 mediates the mechanism of the synergistic activation of the CYP2C9 gene by CAR and HNF4alpha.
Collapse
Affiliation(s)
- Sailesh Surapureddi
- Laboratory of Pharmacology (S.S., R.R., J.A.G.), National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709 and Department of Pathology (J.K.R.), Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Ritu Rana
- Laboratory of Pharmacology (S.S., R.R., J.A.G.), National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709 and Department of Pathology (J.K.R.), Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Janardan K Reddy
- Laboratory of Pharmacology (S.S., R.R., J.A.G.), National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709 and Department of Pathology (J.K.R.), Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Joyce A Goldstein
- Laboratory of Pharmacology (S.S., R.R., J.A.G.), National Institute of Environmental Health Sciences, National Institutes of Health, P.O. Box 12233, Research Triangle Park, NC 27709 and Department of Pathology (J.K.R.), Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611-3008
| |
Collapse
|
44
|
Weber M, Wehling M, Lösel R. Proteins interact with the cytosolic mineralocorticoid receptor depending on the ligand. Am J Physiol Heart Circ Physiol 2008; 295:H361-5. [DOI: 10.1152/ajpheart.00825.2007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Steroid receptors belonging to the superfamily of nuclear receptors do not exist as single monomeric proteins but mediate their effects by the interaction with numerous other proteins, e.g., cofactors for transcription, but also other proteins involved in cellular signaling. This interaction may be ligand dependent, which explains the differential effects of receptor ligands. Whereas some receptors, e.g., the estrogen receptor, have been studied in great detail, much less is known about proteins interacting with the mineralocorticoid receptor (MR). In this study, we aimed to identify interacting proteins using a proteomics approach involving tagged receptor constructs. After affinity isolation of MR complexes, blue native electrophoresis revealed the presence of several populations of MR complexes differing in size and composition. During the identification of interacting proteins, various heat shock proteins but also several previously undescribed potential interactors were found, including 14-3-3-ε. We also demonstrate here that the cytosolic MR in the presence of detergent interacts in a ligand-selective manner with glucose-regulated protein 78 and propionyl-CoA carboxylase-β precursor, which are found in the unliganded or aldosterone-containing complex but not with spironolactone.
Collapse
|
45
|
Antonson P, Jakobsson T, Almlöf T, Guldevall K, Steffensen KR, Gustafsson JÅ. RAP250 Is a Coactivator in the Transforming Growth Factor β Signaling Pathway That Interacts with Smad2 and Smad3. J Biol Chem 2008; 283:8995-9001. [DOI: 10.1074/jbc.m707203200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
46
|
Mahajan MA, Samuels HH. Nuclear receptor coactivator/coregulator NCoA6(NRC) is a pleiotropic coregulator involved in transcription, cell survival, growth and development. NUCLEAR RECEPTOR SIGNALING 2008; 6:e002. [PMID: 18301782 PMCID: PMC2254332 DOI: 10.1621/nrs.06002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 12/11/2007] [Indexed: 11/20/2022]
Abstract
NCoA6 (also referred to as NRC, ASC-2, TRBP, PRIP and RAP250) was originally isolated as a ligand-dependent nuclear receptor interacting protein. However, NCoA6 is a multifunctional coregulator or coactivator necessary for transcriptional activation of a wide spectrum of target genes. The NCoA6 gene is amplified and overexpressed in breast, colon and lung cancers. NCoA6 is a 250 kDa protein which harbors a potent N-terminal activation domain, AD1; and a second, centrally-located activation domain, AD2, which is necessary for nuclear receptor signaling. The intrinsic activation potential of NCoA6 is regulated by its C-terminal STL regulatory domain. Near AD2 is an LxxLL-1 motif which interacts with a wide spectrum of ligand-bound NRs with high-affinity. A second LxxLL motif (LxxLL-2) located towards the C-terminal region is more restricted in its NR specificity. The potential role of NCoA6 as a co-integrator is suggested by its ability to enhance transcriptional activation of a wide variety of transcription factors and from its in vivo association with a number of known cofactors including CBP/p300. NCoA6 has been shown to associate with at least three distinct coactivator complexes containing Set methyltransferases as core polypeptides. The composition of these complexes suggests that NCoA6 may play a fundamental role in transcriptional activation by modulating chromatin structure through histone methylation. Knockout studies in mice suggest that NCoA6 is an essential coactivator. NCoA6-/- embryos die between 8.5-12.5 dpc from general growth retardation coupled with developmental defects in the heart, liver, brain and placenta. NCoA6-/- MEFs grow at a reduced rate compared to WT MEFs and spontaneously undergo apoptosis, indicating the importance of NCoA6 as a prosurvival and anti-apoptotic gene. Studies with NCoA6+/- and conditional knockout mice suggest that NCoA6 is a pleiotropic coregulator involved in growth, development, wound healing and maintenance of energy homeostasis.
Collapse
Affiliation(s)
- Muktar A Mahajan
- Department of Pharmacology, NYU School of Medicine, New York, New York, USA.
| | | |
Collapse
|
47
|
Hao Z, Li X, Qiao T, Fan D. Successful expression and purification of human CIAPIN1 in baculovirus-insect cell system and application of this system to investigation of its potential methyltransferase activity. Int J Biol Macromol 2007; 42:27-32. [PMID: 17935775 DOI: 10.1016/j.ijbiomac.2007.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 08/28/2007] [Accepted: 08/30/2007] [Indexed: 11/24/2022]
Abstract
CIAPIN1 is a newly identified anti-apoptosis molecule which plays an important role in definitive haematopoiesis in mouse fetal liver and confers multidrug resistance in gastric cancer cells. However, the biophysical function of CIAPIN1 is far from elucidated. Bioinformatics predicts that CIAPIN1 may contain a generic methyltransferase motif and a Zn-ribbon-like motif. Based on these data, we postulated that CIAPIN1 might be a DNA or RNA methyltransferase. To substantiate this proposal, recombinant human CIAPIN1 (rhCIAPIN1) was expressed by a baculovirus-insect cell system and purified by Ni-NTA affinity chromatography. In vitro DNA and RNA methyltransferring tests, DNA demethylation test and S-adenosyl-l-[methyl-3H]methionine (3H-AdoMet) binding test were carried out. Our experiments failed to demonstrate that rhCIAPIN1 had any DNA, RNA methyltransferase activity, DNA demethylase activity, or had the capability of binding AdoMet in vitro. Further studies are needed to definitely clarify whether CIAPIN1 has methyltransferase activity.
Collapse
Affiliation(s)
- Zhiming Hao
- Department of Gastroenterology, 1st Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | | | | | | |
Collapse
|
48
|
Sarkar J, Qi C, Guo D, Ahmed MR, Jia Y, Usuda N, Viswakarma N, Rao MS, Reddy JK. Transcription coactivator PRIP, the peroxisome proliferator-activated receptor (PPAR)-interacting protein, is redundant for the function of nuclear receptors PParalpha and CAR, the constitutive androstane receptor, in mouse liver. Gene Expr 2007; 13:255-69. [PMID: 17605299 PMCID: PMC6032459 DOI: 10.3727/000000006780666948] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Disruption of the genes encoding for the transcription coactivators, peroxisome proliferator-activated receptor (PPAR)-interacting protein (PRIP/ASC-2/RAP250/TRBP/NRC) and PPAR-binding protein (PBP/TRAP220/DRIP205/MED1), results in embryonic lethality by affecting placental and multiorgan development. Targeted deletion of coactivator PBP gene in liver parenchymal cells (PBP(LIV-/-)) results in the near abrogation of the induction of PPARalpha and CAR (constitutive androstane receptor)-regulated genes in liver. Here, we show that targeted deletion of coactivator PRIP gene in liver (PRIP(LIV-/-)) does not affect the induction of PPARalpha-regulated pleiotropic responses, including hepatomegaly, hepatic peroxisome proliferation, and induction of mRNAs of genes involved in fatty acid oxidation system, indicating that PRIP is not essential for PPARalpha-mediated transcriptional activity. We also provide additional data to show that liver-specific deletion of PRIP gene does not interfere with the induction of genes regulated by nuclear receptor CAR. Furthermore, disruption of PRIP gene in liver did not alter zoxazolamine-induced paralysis, and acetaminophen-induced hepatotoxicity. Studies with adenovirally driven EGFP-CAR expression in liver demonstrated that, unlike PBP, the absence of PRIP does not prevent phenobarbital-mediated nuclear translocation/retention of the receptor CAR in liver in vivo and cultured hepatocytes in vitro. These results show that PRIP deficiency in liver does not interfere with the function of nuclear receptors PPARalpha and CAR. The dependence of PPARalpha- and CAR-regulated gene transcription on coactivator PBP but not on PRIP attests to the existence of coactivator selectivity in nuclear receptor function.
Collapse
Affiliation(s)
- Joy Sarkar
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CNA, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacol Rev 2006; 58:726-41. [PMID: 17132851 DOI: 10.1124/pr.58.4.5] [Citation(s) in RCA: 734] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The three peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear hormone receptor superfamily. They share a high degree of structural homology with all members of the superfamily, particularly in the DNA-binding domain and ligand- and cofactor-binding domain. Many cellular and systemic roles have been attributed to these receptors, reaching far beyond the stimulation of peroxisome proliferation in rodents after which they were initially named. PPARs exhibit broad, isotype-specific tissue expression patterns. PPARalpha is expressed at high levels in organs with significant catabolism of fatty acids. PPARbeta/delta has the broadest expression pattern, and the levels of expression in certain tissues depend on the extent of cell proliferation and differentiation. PPARgamma is expressed as two isoforms, of which PPARgamma2 is found at high levels in the adipose tissues, whereas PPARgamma1 has a broader expression pattern. Transcriptional regulation by PPARs requires heterodimerization with the retinoid X receptor (RXR). When activated by a ligand, the dimer modulates transcription via binding to a specific DNA sequence element called a peroxisome proliferator response element (PPRE) in the promoter region of target genes. A wide variety of natural or synthetic compounds was identified as PPAR ligands. Among the synthetic ligands, the lipid-lowering drugs, fibrates, and the insulin sensitizers, thiazolidinediones, are PPARalpha and PPARgamma agonists, respectively, which underscores the important role of PPARs as therapeutic targets. Transcriptional control by PPAR/RXR heterodimers also requires interaction with coregulator complexes. Thus, selective action of PPARs in vivo results from the interplay at a given time point between expression levels of each of the three PPAR and RXR isotypes, affinity for a specific promoter PPRE, and ligand and cofactor availabilities.
Collapse
Affiliation(s)
- Liliane Michalik
- Center for Integrative Genomics, National Research Centre "Frontiers in Genetics," University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sun Y, Ding L, Zhang H, Han J, Yang X, Yan J, Zhu Y, Li J, Song H, Ye Q. Potentiation of Smad-mediated transcriptional activation by the RNA-binding protein RBPMS. Nucleic Acids Res 2006; 34:6314-26. [PMID: 17099224 PMCID: PMC1669761 DOI: 10.1093/nar/gkl914] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 10/15/2006] [Accepted: 10/16/2006] [Indexed: 11/25/2022] Open
Abstract
Smad2, Smad3 and Smad4 proteins are considered to be key mediators of transforming growth factor-beta (TGF-beta) signaling. However, the identities of the Smad partners mediating TGF-beta signaling are not fully understood. Here, we show that RNA-binding protein with multiple splicing (RBPMS), a member of the RNA-binding protein family, physically interacts with Smad2, Smad3 and Smad4 both in vitro and in vivo. The presence of TGF-beta increases the binding of RBPMS with these Smad proteins. Consistent with the binding results, overexpression of RBPMS enhances Smad-dependent transcriptional activity in a TGF-beta-dependent manner, whereas knockdown of RBPMS decreases this activity. RBPMS interacts with TGF-beta receptor type I (TbetaR-I), increases phosphorylation of C-terminal SSXS regions in Smad2 and Smad3, and promotes the nuclear accumulation of the Smad proteins. Moreover, RBPMS fails to enhance the transcriptional activity of Smad2 and Smad3 that lack the C-terminal phosphorylation sites. Our data provide the first evidence for an RNA-binding protein playing a role in regulation of Smad-mediated transcriptional activity and suggest that RBPMS stimulates Smad-mediated transactivation possibly through enhanced phosphorylation of Smad2 and Smad3 at the C-terminus and promotion of the nuclear accumulation of the Smad proteins.
Collapse
Affiliation(s)
- Yan Sun
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Lihua Ding
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Hao Zhang
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Juqiang Han
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Xiao Yang
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Jinghua Yan
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Yunfeng Zhu
- The 307 Hospital, Beijing 100071People's Republic of China
| | - Jiezhi Li
- Beijing Institute of Biotechnology, Beijing 100850People's Republic of China
- The 307 Hospital, Beijing 100071People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Haifeng Song
- Beijing Institute of Radiation Medicine, Beijing 100850People's Republic of China
| | - Qinong Ye
- To whom correspondence should be addressed. Tel: +8610 6818 0809; Fax: +8610 6824 8045;
| |
Collapse
|