99901
|
Song G, Yang L. Inhibited CD47 gene affects the clearance of acute myelogenous leukemia stem cells. J Cell Biochem 2018; 120:10303-10309. [PMID: 30565723 DOI: 10.1002/jcb.28314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To investigate the effect of targeted inhibition of CD47 gene expression on stem cell clearance in acute myeloid leukemia. METHODS After the lentiviral CD47-siRNA was transfected into acute myelogenous leukemia stem cells (LSCs), the proliferative status of acute myelogenous LSCs was detected by cell counting kit-8, and the apoptosis of stem cells of acute myeloid leukemia was detected by annexin/propidium iodide flow assays. The expression of Bcl-2, Bcl-xl, MCL-1, PIK3p110β, and interleukin (IL)-3 in acute myeloid LSCs was detected by Western blot analysis and the activity of protein phosphatase 2A (PP2A) and the protein content of CD96 and CD90 were measured by enzyme-linked immunosorbent assay kits. RESULTS After transfection of the lentivirus CD47-siRNA into acute myeloid LSCs, compared with the empty vector transfection group (control group), the cell viability of the CD47-siRNA transfection group was decreased, and the apoptosis rate was increased. Furthermore, the antiapoptotic protein Bcl-2, Bcl-xl, and MCL-1 and the content of IL-3 protein, CD96, and CD90 was decreased, whereas the activity of PIK3p110β and PP2A protein was increased. CONCLUSION Targeted inhibition of CD47 could inhibit the proliferation of myeloid LSCs, promote apoptosis, mobilize the cells into the cell cycle, and reduce the high expression of immune proteins on the cell surface, therefore providing a theoretical basis for the elimination and eradication of LSCs.
Collapse
Affiliation(s)
- Guangle Song
- Morphological Laboratory, Taishan Medical College, Taian, Shandong, China
| | - Leiying Yang
- Department of Pathology, Taishan Medical College, Taian, Shandong, China
| |
Collapse
|
99902
|
Analysis of genes encoding seed storage proteins (SSPs) in chickpea (Cicer arietinum L.) reveals co-expressing transcription factors and a seed-specific promoter. Funct Integr Genomics 2018; 19:373-390. [PMID: 30560463 DOI: 10.1007/s10142-018-0650-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 12/27/2022]
Abstract
Improvement of the quality and quantity of chickpea seed protein can be greatly facilitated by an understanding of the genic organization and the genetic architecture of the genes encoding seed storage proteins (SSPs). The aim of this study was to provide a comprehensive analysis of the chickpea SSP genes, putative co-expressing transcription factors (TFs), and to identify a seed-specific SSP gene promoter. A genome-wide identification of SSP genes in chickpea led to the identification of 21 non-redundant SSP encoding genes located on 6 chromosomes. Phylogenetic analysis grouped SSP genes into 3 subgroups where members within the same clade demonstrated similar motif composition and intron-exon organization. Tandem duplications were identified to be the major contributors to the expansion of the SSP gene family in chickpea. Co-expression analysis revealed 14 TFs having expression profiles similar to the SSP genes that included members of important TF families that are known to regulate seed development. Expression analysis of SSP genes and TFs revealed significantly higher expression in late stages of seed development as well as in high seed protein content (HPC) genotypes. In silico analysis of the promoter regions of the SSP encoding genes revealed several seed-specific cis-regulatory elements such as RY repeats, ACGT motifs, CAANTG, and GCN4. A candidate promoter was analyzed for seed specificity by generating stable transgenics in Arabidopsis. Overall, this study provides a useful resource to explore the regulatory networks involved in SSP synthesis and/or accumulation for utilization in developing nutritionally improved chickpea genotypes.
Collapse
|
99903
|
Gong YN, Crawford JC, Heckmann BL, Green DR. To the edge of cell death and back. FEBS J 2018; 286:430-440. [PMID: 30506628 DOI: 10.1111/febs.14714] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/28/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Programmed cell death plays a central role in maintaining homeostasis. Various studies have demonstrated that programmed cell death is not a one-way street; cells can survive even when the core cell death processes are underway. Cell death initiation, prevention, and recovery function in a coordinated fashion to establish and maintain a homeostatic environment. In this review, we discuss how dying cells can be rescued from death's grip and the subsequent physiological consequences. We suggest a fundamental question to be answered-at least at the single cell level is, can we predict if a certain cell is more or less likely to survive or die? And importantly, what physiological and pathological consequences, as well as therapeutic approaches can we delineate from this ability to predict cell death versus survival.
Collapse
Affiliation(s)
- Yi-Nan Gong
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
99904
|
Zong X, Zhao J, Wang H, Lu Z, Wang F, Du H, Wang Y. Mettl3 Deficiency Sustains Long-Chain Fatty Acid Absorption through Suppressing Traf6-Dependent Inflammation Response. THE JOURNAL OF IMMUNOLOGY 2018; 202:567-578. [PMID: 30567729 DOI: 10.4049/jimmunol.1801151] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023]
Abstract
A better understanding of the molecular mechanism of intestinal fatty acid absorption could lead to novel approaches to treatment and prevention of fatty acid-related metabolic diseases. Although it is confirmed that absorption of long-chain fatty acids (LCFAs) decreases during the pathological processes, the genetic basis and molecular mechanisms remain largely unknown. N 6-methyladenosine (m6A) is the most prevalent internal modification on eukaryotic mRNA. Recently, m6A has been found to play important roles in inflammation and antiviral responses. In this study, we show that deficiency of Mettl3, the core methyltransferase of m6A, exerts antimalabsorption of LCFA activity in vitro through inhibiting the inflammation response mediated by LPS. To substantiate this finding further, we found the levels of triglycerides were also sustained in cells with depleted Mettl3, which were cultured in Transwell to polarize with villus formation to simulate the situation in vivo. Mechanistically, depletion of Mettl3 decreases the m6A level of Traf6 mRNA, thereby its transcripts are entrapped in the nucleus, followed by the decreased expression of Traf6, leading to the suppression of NF-κB and MAPK signaling pathway. Thus, the inflammation response was suppressed, resulting in the sustained absorption of LCFA. Moreover, we found that ectopic expression of Traf6 largely abolishes the sustained absorption LCFA in Mettl3 depletion cells. Collectively, silencing Mettl3 could sustain LCFA absorption through blocking the TRAF6-dependent inflammation response. Our work uncovers a critical function of m6A methylation and provides insight into critical roles of Mettl3 in LCFA absorption and inflammatory disease.
Collapse
Affiliation(s)
- Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Jing Zhao
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Hong Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and
| | - Zeqing Lu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Fengqin Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Huahua Du
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province 310058, People's Republic of China; and .,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, Hangzhou, Zhejiang Province 310058, People's Republic of China
| |
Collapse
|
99905
|
Wang G, Fu XL, Wang JJ, Guan R, Sun Y, Tony To SS. Inhibition of glycolytic metabolism in glioblastoma cells by Pt3glc combinated with PI3K inhibitor via SIRT3‐mediated mitochondrial and PI3K/Akt–MAPK pathway. J Cell Physiol 2018; 234:5888-5903. [PMID: 29336479 DOI: 10.1002/jcp.26474] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/27/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Xing-Li Fu
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
- Information Central, Hubei University of Medicine, Shiyan, China
| | - Rui Guan
- Information Central, Hubei University of Medicine, Shiyan, China
| | - Yan Sun
- Information Central, Hubei University of Medicine, Shiyan, China
| | - Shing-Shun Tony To
- Department of Healthy Technology and Information, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| |
Collapse
|
99906
|
Denton D, Kumar S. Autophagy-dependent cell death. Cell Death Differ 2018; 26:605-616. [PMID: 30568239 DOI: 10.1038/s41418-018-0252-y] [Citation(s) in RCA: 503] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy-dependent cell death can be defined as cell demise that has a strict requirement of autophagy. Although autophagy often accompanies cell death following many toxic insults, the requirement of autophagic machinery for cell death execution, as established through specific genetic or chemical inhibition of the process, is highly contextual. During animal development, perhaps the best validated model of autophagy-dependent cell death is the degradation of the larval midgut during larval-pupal metamorphosis, where a number of key autophagy genes are required for the removal of the tissues. Surprisingly though, even in the midgut, not all of the 'canonical' autophagic machinery appears to be required. In other organisms and cancer cells many variations of autophagy-dependent cell death are apparent, pointing to the lack of a unifying cell death pathway. It is thus possible that components of the autophagy machinery are selectively utilised or repurposed for this type of cell death. In this review, we discuss examples of cell death that utilise autophagy machinery (or part thereof), the current knowledge of the complexity of autophagy-dependent cellular demise and the potential mechanisms and regulatory pathways involved in such cell death.
Collapse
Affiliation(s)
- Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| |
Collapse
|
99907
|
Li X, Jin Q, Zhang Y, Wu YL, Jin CM, Cui BW, Li Y, Jin MJ, Shang Y, Jiang M, Yang HX, Wu M, Liu J, Lian LH, Nan JX. Inhibition of P2X7R-NLRP3 Inflammasome Activation by Pleurotus citrinopileatus: A Possible Protective Role in Alcoholic Hepatosteatosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:13183-13190. [PMID: 30497264 DOI: 10.1021/acs.jafc.8b05756] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Pleurotus citrinopileatus (golden oyster mushroom) is a widely used edible mushroom. We investigated the inhibitory effect of P. citrinopileatus aqueous extract against alcoholic steatohepatitis and its underlying mechanism. Acute and chronic ethanol-feeding murine models were established by intragastrically administering ethanol or feeding an ethanol-containing Lieber-DeCarli liquid diet to male C57BL/6 mice. In both models, P. citrinopileatus decreased serum alanine aminotransferase (ALT), aspartate transaminase (AST), triglyceride (TG), and hepatic TG levels. Hematoxylin and eosin (HE) and Oil Red O staining confirmed that P. citrinopileatus ameliorated both acute and chronic alcoholic hepatosteatosis, characterized by regulation of lipid-metabolism-related proteins, including sirtuin 1 (SIRT1), AMP-activated kinase (AMPK), and sterol regulatory element-binding protein (SREBP1). P. citrinopileatus reversed inflammatory response via modulating purinergic receptor P2X ligand-gated ion channel 7 (P2X7R)-NOD-like receptor pyrin domain 3 (NLRP3) inflammasome. P. citrinopileatus restored the expressions of those proteins to a normal level. In addition, HepG2 cells were incubated with P. citrinopileatus prior to ethanol stimulation. P. citrinopileatus reduced ethanol exposure-induced lipid deposition. Concomitantly, P. citrinopileatus increased AMPK and SIRT1 expressions, which were reduced by ethanol treatment. P. citrinopileatus ameliorated alcoholic hepatic steatosis and accompanied inflammatory response via regulating SIRT1-AMPK and P2X7R-NLRP3 inflammasome activation, highlighting a promising strategy and utility of P. citrinopileatus for alcoholic steatohepatitis as dietary health supplements.
Collapse
Affiliation(s)
- Xia Li
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Quan Jin
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Yu Zhang
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Yan-Ling Wu
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Cheng-Min Jin
- Analysis Center , Dt&CRO, Incorporated , Yongin-si , Gyeonggi-do 17042 , Republic of Korea
| | - Ben-Wen Cui
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Ying Li
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Ming-Ji Jin
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Yue Shang
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Min Jiang
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Hong-Xu Yang
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Mei Wu
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Jian Liu
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Li-Hua Lian
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
| | - Ji-Xing Nan
- Key Laboratory for Natural Resource of Changbai Mountain and Functional Molecules, Ministry of Education, College of Pharmacy , Yanbian University , Yanji , Jilin 133002 , People's Republic of China
- Clinical Research Center , Yanbian University Hospital , Yanji , Jilin 133002 , People's Republic of China
| |
Collapse
|
99908
|
Holt ME, Salay LE, O’Brien E, Barton JK, Chazin WJ. Functional and structural similarity of human DNA primase [4Fe4S] cluster domain constructs. PLoS One 2018; 13:e0209345. [PMID: 30562384 PMCID: PMC6298731 DOI: 10.1371/journal.pone.0209345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/14/2018] [Indexed: 01/15/2023] Open
Abstract
The regulatory subunit of human DNA primase has a C-terminal domain (p58C) that contains a [4Fe4S] cluster and binds DNA. Previous electrochemical analysis of a p58C construct revealed that its affinity for DNA is sensitive to the redox state of the [4Fe4S] cluster. Concerns about the validity of this conclusion have been raised, based in part on differences in X-ray crystal structures of the p58C272-464 construct used for that study and that of a N-terminally shifted p58C266-456 construct and consequently, an assumption that p58C272-464 has abnormal physical and functional properties. To address this controversy, a new p58C266-464 construct containing all residues was crystallized under the conditions previously used for crystallizing p58C272-464, and the solution structures of both constructs were assessed using circular dichroism and NMR spectroscopy. In the new crystal structure, p58C266-464 exhibits the same elements of secondary structure near the DNA binding site as observed in the crystal structure of p58C272-464. Moreover, in solution, circular dichroism and 15N,1H-heteronuclear single quantum coherence (HSQC) NMR spectra show there are no significant differences in the distribution of secondary structures or in the tertiary structure or the two constructs. To validate that the two constructs have the same functional properties, binding of a primed DNA template was measured using a fluorescence-based DNA binding assay, and the affinities for this substrate were the same (3.4 ± 0.5 μM and 2.7 ± 0.3 μM, respectively). The electrochemical properties of p58C266-464 were also measured and this p58C construct was able to engage in redox switching on DNA with the same efficiency as p58C272-464. Together, these results show that although p58C can be stabilized in different conformations in the crystalline state, in solution there is effectively no difference in the structure and functional properties of p58C constructs of different lengths.
Collapse
Affiliation(s)
- Marilyn E. Holt
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Lauren E. Salay
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elizabeth O’Brien
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Jacqueline K. Barton
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Walter J. Chazin
- Departments of Biochemistry and Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
99909
|
Kaur H, Sain N, Mohanty D, Salunke DM. Deciphering evolution of immune recognition in antibodies. BMC STRUCTURAL BIOLOGY 2018; 18:19. [PMID: 30563492 PMCID: PMC6299584 DOI: 10.1186/s12900-018-0096-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/14/2018] [Indexed: 11/29/2022]
Abstract
Background Antibody, the primary effector molecule of the immune system, evolves after initial encounter with the antigen from a precursor form to a mature one to effectively deal with the antigen. Antibodies of a lineage diverge through antigen-directed isolated pathways of maturation to exhibit distinct recognition potential. In the context of evolution in immune recognition, diversity of antigen cannot be ignored. While there are reports on antibody lineage, structural perspective with respect to diverse recognition potential in a lineage has never been studied. Hence, it is crucial to evaluate how maturation leads to topological tailoring within a lineage enabling them to interact with significantly distinct antigens. Results A data-driven approach was undertaken for the study. Global experimental mouse and human antibody-antigen complex structures from PDB were compiled into a coherent database of germline-linked antibodies bound with distinct antigens. Structural analysis of all lineages showed variations in CDRs of both H and L chains. Observations of conformational adaptation made from analysis of static structures were further evaluated by characterizing dynamics of interaction in two lineages, mouse VH1–84 and human VH5–51. Sequence and structure analysis of the lineages explained that somatic mutations altered the geometries of individual antibodies with common structural constraints in some CDRs. Additionally, conformational landscape obtained from molecular dynamics simulations revealed that incoming pathogen led to further conformational divergence in the paratope (as observed across datasets) even while maintaining similar overall backbone topology. MM-GB/SA analysis showed binding energies to be in physiological range. Results of the study are coherent with experimental observations. Conclusions The findings of this study highlight basic structural principles shaping the molecular evolution of a lineage for significantly diverse antigens. Antibodies of a lineage follow different developmental pathways while preserving the imprint of the germline. From the study, it can be generalized that structural diversification of the paratope is an outcome of natural selection of a conformation from an available ensemble, which is further optimized for antigen interaction. The study establishes that starting from a common lineage, antibodies can mature to recognize a wide range of antigens. This hypothesis can be further tested and validated experimentally. Electronic supplementary material The online version of this article (10.1186/s12900-018-0096-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Harmeet Kaur
- Regional Centre for Biotechnology, Biotech Science Cluster, Faridabad, Haryana, 121001, India.,Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Neetu Sain
- National Institute of Immunology, New Delhi, Delhi, 110067, India
| | - Debasisa Mohanty
- National Institute of Immunology, New Delhi, Delhi, 110067, India
| | - Dinakar M Salunke
- Regional Centre for Biotechnology, Biotech Science Cluster, Faridabad, Haryana, 121001, India. .,International Centre for Genetic Engineering and Biotechnology, New Delhi, Delhi, 110067, India.
| |
Collapse
|
99910
|
HDAC1 overexpression enhances β-cell proliferation by down-regulating Cdkn1b/p27. Biochem J 2018; 475:3997-4010. [PMID: 30322885 DOI: 10.1042/bcj20180465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/12/2018] [Accepted: 10/14/2018] [Indexed: 12/18/2022]
Abstract
The homeobox transcription factor Nkx6.1 is sufficient to increase functional β-cell mass, where functional β-cell mass refers to the combination of β-cell proliferation, glucose-stimulated insulin secretion (GSIS) and β-cell survival. Here, we demonstrate that the histone deacetylase 1 (HDAC1), which is an early target of Nkx6.1, is sufficient to increase functional β-cell mass. We show that HDAC activity is necessary for Nkx6.1-mediated proliferation, and that HDAC1 is sufficient to increase β-cell proliferation in primary rat islets and the INS-1 832/13 β-cell line. The increase in HDAC1-mediated proliferation occurs while maintaining GSIS and increasing β-cell survival in response to apoptotic stimuli. We demonstrate that HDAC1 overexpression results in decreased expression of the cell cycle inhibitor Cdkn1b/p27 which is essential for inhibiting the G1 to S phase transition of the cell cycle. This corresponds with increased expression of key cell cycle activators, such as Cyclin A2, Cyclin B1 and E2F1, which are activated by activation of the Cdk4/Cdk6/Cyclin D holoenzymes due to down-regulation of Cdkn1b/p27. Finally, we demonstrate that overexpression of Cdkn1b/p27 inhibits HDAC1-mediated β-cell proliferation. Our data suggest that HDAC1 is critical for the Nkx6.1-mediated pathway that enhances functional β-cell mass.
Collapse
|
99911
|
Abstract
Ribosomes, which synthesize the proteins of a cell, comprise ribosomal RNA and ribosomal proteins, which coassemble hierarchically during a process termed ribosome biogenesis. Historically, biochemical and molecular biology approaches have revealed how preribosomal particles form and mature in consecutive steps, starting in the nucleolus and terminating after nuclear export into the cytoplasm. However, only recently, due to the revolution in cryo-electron microscopy, could pseudoatomic structures of different preribosomal particles be obtained. Together with in vitro maturation assays, these findings shed light on how nascent ribosomes progress stepwise along a dynamic biogenesis pathway. Preribosomes assemble gradually, chaperoned by a myriad of assembly factors and small nucleolar RNAs, before they reach maturity and enter translation. This information will lead to a better understanding of how ribosome synthesis is linked to other cellular pathways in humans and how it can cause diseases, including cancer, if disturbed.
Collapse
Affiliation(s)
- Jochen Baßler
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany; ,
| | - Ed Hurt
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany; ,
| |
Collapse
|
99912
|
Rout AK, Mishra J, Dehury B, Maharana J, Acharya V, Karna SK, Parida PK, Behera BK, Das BK. Structural bioinformatics insights into ATP binding mechanism in zebrafish (Danio rerio) cyclin-dependent kinase-like 5 (zCDKL5) protein. J Cell Biochem 2018; 120:9437-9447. [PMID: 30569538 DOI: 10.1002/jcb.28219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/15/2018] [Indexed: 11/06/2022]
Abstract
In mammalian systems, the conserved cyclin-dependent protein kinases (CDKs) control the process of cell division and curb the transcription mechanism in response to diverse signaling events that are essential for the catalytic activity. In zebrafish, zCDKL5 portrays differential expression profiling in several tissues and presumed to play a vital role in the neuronal development. In this present study, the sequence-structure relationship and mode of ATP binding in zCDKL5 was unveiled through theoretical modeling, molecular docking, and MD simulations. Like human CDKs, the modeled zCDKL5 was found to be bipartite in nature, where, ATP binds to the central cavity of the catalytic domain through a strong network of H-bonding, electrostatic, and hydrophobic interactions. MD simulation portrayed that conserved residues, viz, Ile10, Gly11, Glu12, Val18, Val64, Glu81, Cys143, and Asp144 were indispensable for tight anchoring of ATP and contribute to the stability of the zCDKL5-ATP complex. MM/PBSA binding free energy analysis displayed that van der Waal energy (ΔG vwd ) and Electrostatic energy (ΔG ele ) were the major contributors towards the overall binding free energy. Thus, the comparative structural bioinformatics approach has shed new insights into the dynamics and ATP binding mechanism of zCDKL5. The results from the study will help to undertake further research on the role of phosphorylated CDKL5 in the onset of neurodevelopmental disorders caused by mutations in higher eukaryotic systems.
Collapse
Affiliation(s)
- Ajaya Kumar Rout
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Jagruti Mishra
- Department of Bioinformatics, Orissa University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Budheswar Dehury
- Biomedical Informatics Centre, ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India.,Department of Chemistry, Technical University of Denmark, Denmark
| | - Jitendra Maharana
- Department of Bioinformatics, Orissa University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Varsha Acharya
- Department of Bioinformatics, Orissa University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Subodha Kumar Karna
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Pranaya Kumar Parida
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Bijay Kumar Behera
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Basanta Kumar Das
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| |
Collapse
|
99913
|
The Host Scaffolding Protein Filamin A and the Exocyst Complex Control Exocytosis during InlB-Mediated Entry of Listeria monocytogenes. Infect Immun 2018; 87:IAI.00689-18. [PMID: 30348826 DOI: 10.1128/iai.00689-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Listeria monocytogenes is a foodborne bacterium that causes gastroenteritis, meningitis, or abortion. Listeria induces its internalization (entry) into some human cells through interaction of the bacterial surface protein InlB with its host receptor, the Met tyrosine kinase. InlB and Met promote entry, in part, through stimulation of localized exocytosis. How exocytosis is upregulated during entry is not understood. Here, we show that the human signaling proteins mTOR, protein kinase C-α (PKC-α), and RalA promote exocytosis during entry by controlling the scaffolding protein Filamin A (FlnA). InlB-mediated uptake was accompanied by PKC-α-dependent phosphorylation of serine 2152 in FlnA. Depletion of FlnA by RNA interference (RNAi) or expression of a mutated FlnA protein defective in phosphorylation impaired InlB-dependent internalization. These findings indicate that phosphorylation of FlnA by PKC-α contributes to entry. mTOR and RalA were found to mediate the recruitment of FlnA to sites of InlB-mediated entry. Depletion of PKC-α, mTOR, or FlnA each reduced exocytosis during InlB-mediated uptake. Because the exocyst complex is known to mediate polarized exocytosis, we examined if PKC-α, mTOR, RalA, or FlnA affects this complex. Depletion of PKC-α, mTOR, RalA, or FlnA impaired recruitment of the exocyst component Exo70 to sites of InlB-mediated entry. Experiments involving knockdown of Exo70 or other exocyst proteins demonstrated an important role for the exocyst complex in uptake of Listeria Collectively, our results indicate that PKC-α, mTOR, RalA, and FlnA comprise a signaling pathway that mobilizes the exocyst complex to promote infection by Listeria.
Collapse
|
99914
|
Pentimalli F, Grelli S, Di Daniele N, Melino G, Amelio I. Cell death pathologies: targeting death pathways and the immune system for cancer therapy. Genes Immun 2018; 20:539-554. [PMID: 30563970 PMCID: PMC6451632 DOI: 10.1038/s41435-018-0052-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/25/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
Alterations in the molecular mechanisms of cell death are a common feature of cancer. These alterations enable malignant cells to survive intrinsic death signalling leading to accumulation of genetic aberrations and helping them to cope with adverse conditions. Regulated cell death has historically been exclusively associated with classical apoptosis; however, increasing evidence indicates that several alternative mechanisms orchestrate multiple death pathways, such as ferroptosis, entosis, necroptosis and immunogenic cell death, each with distinct underlying molecular mechanisms. Although pharmacological targeting of cell death pathways has been the subject of intensive efforts in recent decades with a dominant focus on targeting apoptosis, the identification of these novel death pathways has opened additional venues for intervention in cancer cells and the immune system. In this mini-review, we cover some recent progress on major recently emerged cell death modalities, emphasizing their potential clinical and therapeutic implications. We also discuss the interplay between cell death and immune response, highlighting the potential of the combination of traditional anticancer therapy and immunocheckpoint blockade. While attempting to stimulate discussion and draw attention to the possible clinical impact of these more recently emerged cell death modalities, we also cover the major progress achieved in translating strategies for manipulation of apoptotic pathways into the clinic, focusing on the attempts to target the anti-apoptotic protein BCL-2 and the tumour suppressor p53.
Collapse
Affiliation(s)
- Francesca Pentimalli
- Centro Ricerche Oncologiche Mercogliano (CROM), Istituto Nazionale Tumori - IRCCS -Fondazione G. Pascale, Naples, Italy
| | - Sandro Grelli
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Nicola Di Daniele
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Gerry Melino
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK
| | - Ivano Amelio
- Medical Research Council, Toxicology Unit, University of Cambridge, Hodgkin Building, Lancaster Road, P.O. Box 138, Leicester, LE1 9HN, UK.
| |
Collapse
|
99915
|
ArunSundar M, Shanmugarajan TS, Ravichandiran V. 3,4-Dihydroxyphenylethanol Assuages Cognitive Impulsivity in Alzheimer's Disease by Attuning HPA-Axis via Differential Crosstalk of α7 nAChR with MicroRNA-124 and HDAC6. ACS Chem Neurosci 2018; 9:2904-2916. [PMID: 29901389 DOI: 10.1021/acschemneuro.7b00532] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cognitive impulsivity, a form of suboptimal cost-benefit decision making, is an illustrious attribute of an array of neurodegenerative diseases including Alzheimer's disease (AD). In this study, a delay discounting paradigm was used to assess the effect of 3,4-dihydroxyphenylethanol (DOPET) on cognitive impulsivity, in an oA42i (oligomeric amyloid β1-42 plus ibotenic acid) induced AD mouse model, using a nonspatial T-maze task. The results depicted that oA42i administration elevated cognitive impulsivity, whereas DOPET treatment attenuated the impulsive behavior and matched the choice of the sham-operated controls. In addition, DOPET treatment has ameliorated the anxiety-like behavior in the oA42i-challenged mice. Probing the molecular signaling cascades underpinning these functional ramifications in the oA42i-challenged mice revealed reduced cholinergic (α7 nAChR; alpha 7 nicotinic acetylcholine receptor) function, dysregulated hypothalamic-pituitary-adrenal (HPA) axis (manifested by amplified glucocorticoid receptor expression and plasma corticosterone levels), and also aberrations in the neuroepigenetic (microRNA-124, HDAC6 (histone deacetylase 6), and HSP90 (heat-shock protein 90) expressions) as well as nucleocytoplasmic (importin-α1 expression and nuclear ultra-architecture) continuum. Nonetheless, DOPET administration ameliorated these perturbations and the observations were in line with that of the sham-operated mice. Further validation of the results with organotypic hippocampal slice cultures (OHSCs) confirmed the in vivo findings. We opine that HPA-axis attunement by DOPET might be orchestrated through the α7 nAChR-mediated pathway. Based on these outcomes, we posit that 3,4-dihydroxyphenylethanol might be a potential multimodal agent for the management of cognitive impulsivity and neuromolecular quagmire in AD.
Collapse
Affiliation(s)
- Mohanasundaram ArunSundar
- Department of Pharmacology, School of Pharmaceutical Sciences, Vels University (VISTAS), Pallavaram, Chennai-600117, India
| | | | | |
Collapse
|
99916
|
Duong VT, Unhelkar MH, Kelly JE, Kim SH, Butts CT, Martin RW. Protein structure networks provide insight into active site flexibility in esterase/lipases from the carnivorous plant Drosera capensis. Integr Biol (Camb) 2018; 10:768-779. [PMID: 30516771 PMCID: PMC6336102 DOI: 10.1039/c8ib00140e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In plants, esterase/lipases perform transesterification reactions, playing an important role in the synthesis of useful molecules, such as those comprising the waxy coatings of leaf surfaces. Plant genomes and transcriptomes have provided a wealth of data about expression patterns and the circumstances under which these enzymes are upregulated, e.g. pathogen defense and response to drought; however, predicting their functional characteristics from genomic or transcriptome data is challenging due to weak sequence conservation among the diverse members of this group. Although functional sequence blocks mediating enzyme activity have been identified, progress to date has been hampered by the paucity of information on the structural relationships among these regions and how they affect substrate specificity. Here we present methodology for predicting overall protein flexibility and active site flexibility based on molecular modeling and analysis of protein structure networks (PSNs). We define two new types of specialized PSNs: sequence region networks (SRNs) and active site networks (ASNs), which provide parsimonious representations of molecular structure in reference to known features of interest. Our approach, intended as an aid to target selection for poorly characterized enzyme classes, is demonstrated for 26 previously uncharacterized esterase/lipases from the genome of the carnivorous plant Drosera capensis and validated using a case/control design. Analysis of the network relationships among functional blocks and among the chemical moieties making up the catalytic triad reveals potentially functionally significant differences that are not apparent from sequence analysis alone.
Collapse
Affiliation(s)
- Vy T. Duong
- Department of Chemistry, UC Irvine
- Department of Molecular Biology & Biochemistry, UC Irvine
| | | | | | | | - Carter T. Butts
- Departments of Sociology, Statistics, and Electrical Engineering & Computer Science, UC Irvine
| | - Rachel W. Martin
- Department of Chemistry, UC Irvine
- Department of Molecular Biology & Biochemistry, UC Irvine
| |
Collapse
|
99917
|
Amiri S, Jemmali B, Ferchichi M, Jeljeli H, Boulbaba R, Ben Gara A. Assessment of growth hormone gene polymorphism effects on reproductive traits in Holstein dairy cattle in Tunisia. Arch Anim Breed 2018; 61:481-489. [PMID: 32175457 PMCID: PMC7065384 DOI: 10.5194/aab-61-481-2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/29/2018] [Indexed: 11/11/2022] Open
Abstract
Research to assess the effect of single genes on reproductive traits in bovine species is imperative to elucidate genes' functions and acquire a better perspective of quantitative traits. The present study was undertaken to characterize genetic diversity in the bovine growth hormone (GH) gene in a population of 410 Holstein dairy cows in Tunisia. The analyses were based on single nucleotide polymorphisms, and GH-AluI and GH-MspI detections and genotyping were carried out using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Data were analyzed using a mixed linear model with the MIXED procedure to reveal the possible effect of GH genotypes on reproductive traits. The frequency data of AluI(L//V) and MspI( + // - ) alleles were 87.04//12.96 and 70.06//29.94, respectively. The distribution of the frequency of GH genotypes for LL/LV/VV and ( - / - )//( + / - )//( + / + ) were 77.75//18.59//3.66 and 15.37//29.13//55.50, respectively. The results of the statistical analyses proved that GH-AluI showed a substantial favorable effect on exanimate traits except for the age at first calving; however, only a suggestive effect of GH-MspI on the calving interval (CI) and the days open (DI) was found. The homozygous LL genotype seemed to be advantageous with respect to the CI and the DI compared with LV and VV genotypes. Heterozygous MspI( + / - ) cows tended to have a longer CI and DI than MspI( + / + ) and MspI( - / - ) cows, but the difference was not statistically significant. A significant effect of different GH-AluI-MspI combined genotypes was found on the number of inseminations per conception, the CI and the DI, and the LL/- combined genotype seemed to be associated with better reproductive performance. Based on these results, the LL genotype of the GH locus can be considered to be a favorable genotype for reproductive traits in Holstein dairy cattle, although these findings need to be confirmed by further research before polymorphisms can be used in a marker-assisted selection program.
Collapse
Affiliation(s)
- Sihem Amiri
- National Agronomic Institute of Tunisia, 43 Charles Nicoles street 1082, Tunis-Mahrajène, Tunisia
| | - Bayrem Jemmali
- Laboratory of Improvement and Integrated Development of Animal Productivity and Food Resources,
Higher School of Agriculture of Mateur, University of Carthage, Tunis, Tunisia
| | - Mohamed Amine Ferchichi
- National Agronomic Institute of Tunisia, 43 Charles Nicoles street 1082, Tunis-Mahrajène, Tunisia
| | - Hajer Jeljeli
- Laboratory of Improvement and Integrated Development of Animal Productivity and Food Resources,
Higher School of Agriculture of Mateur, University of Carthage, Tunis, Tunisia
| | - Rekik Boulbaba
- Laboratory of Improvement and Integrated Development of Animal Productivity and Food Resources,
Higher School of Agriculture of Mateur, University of Carthage, Tunis, Tunisia
| | - Abderrahmane Ben Gara
- Laboratory of Improvement and Integrated Development of Animal Productivity and Food Resources,
Higher School of Agriculture of Mateur, University of Carthage, Tunis, Tunisia
| |
Collapse
|
99918
|
Zhang X, Yuan Y, Wang L, Guo Y, Li M, Li C, Pu X. Use multiscale simulation to explore the effects of the homodimerizations between different conformation states on the activation and allosteric pathway for the μ-opioid receptor. Phys Chem Chem Phys 2018; 20:13485-13496. [PMID: 29726867 DOI: 10.1039/c8cp02016g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recently, oligomers of G-protein coupled receptors (GPCRs) have been an important topic in the GPCR fields. However, knowledge about their structures and activation mechanisms is very limited due to the absence of crystal structures reported. In this work, we used multiscale simulations to study the effects of homodimerization between different conformation states on their activation, dynamic behaviors, and allosteric communication pathways for μ-OR. The results indicated that the dimerization of one inactive monomer with either one inactive monomer or one active one could enhance its constitutive activation. However, the conformation state of the other protomer (e.g., active or inactive) can influence the activated extent. The dimerization between the two inactive protomers leads to a negative cooperativity for their activation, which should contribute to the asymmetric activation of GPCR dimers observed in some experiments. On the other hand, for the active monomer, its dimerization with one inactive receptor could alleviate its deactivation, whereby negative and positive cooperativities can be observed between the two subunits of the dimer, depending on the different regions. Observations from protein structure network (PSN) analysis indicated that the dimerization of one inactive monomer with one active one would cause a significant drop in the number of main pathways from the ligand binding pocket to the G-protein coupled region for the inactive protomer, while the impact is minor for the active protomer. But, for the active monomer or the inactive one, its dimerization with one inactive monomer would significantly change the types of residues participating in the pathway with the highest frequency.
Collapse
Affiliation(s)
- Xi Zhang
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, People's Republic of China.
| | - Yuan Yuan
- College of Management, Southwest University for Nationalities, Chengdu 610041, P. R. China
| | - Longrong Wang
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, People's Republic of China.
| | - Yanzhi Guo
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, People's Republic of China.
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, People's Republic of China.
| | - Chuan Li
- College of Computer Science, Sichuan University, Chengdu, Sichuan 610064, P. R. China.
| | - Xuemei Pu
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, People's Republic of China.
| |
Collapse
|
99919
|
Localized protein biotinylation at DNA damage sites identifies ZPET, a repressor of homologous recombination. Genes Dev 2018; 33:75-89. [PMID: 30567999 PMCID: PMC6317314 DOI: 10.1101/gad.315978.118] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Here, Moquin et al. show that fusion of the promiscuous biotin ligase BirAR118G with RAD18 leads to localized protein biotinylation at DNA damage sites and identify ZPET/ZNF280C as a potential DNA damage response protein. Their findings show that ZPET is an HR repressor and also suggest that localized protein biotinylation at DNA damage sites is a useful strategy to identify DDR proteins. Numerous DNA repair and signaling proteins function at DNA damage sites to protect the genome. Here, we show that fusion of the promiscuous biotin ligase BirAR118G with RAD18 leads to localized protein biotinylation at DNA damage sites, allowing identification of ZPET (zinc finger protein proximal to RAD eighteen)/ZNF280C as a potential DNA damage response (DDR) protein. ZPET binds ssDNA and localizes to DNA double-strand breaks (DSBs) and stalled replication forks. In vitro, ZPET inhibits MRE11 binding to ssDNA. In cells, ZPET delays MRE11 binding to chromatin after DSB formation and slows DNA end resection through binding ssDNA. ZPET hinders resection independently of 53BP1 and HELB. Cells lacking ZPET displayed enhanced homologous recombination (HR), accelerated replication forks under stress, and increased resistance to DSBs and PARP inhibition. These results not only reveal ZPET as an HR repressor but also suggest that localized protein biotinylation at DNA damage sites is a useful strategy to identify DDR proteins.
Collapse
|
99920
|
Zhang M, Deng X, Guan X, Geng L, Fu M, Zhang B, Chen R, Hu H, Hu K, Zhang D, Li M, Liu Y, Gong S, Hu Q. Herpes Simplex Virus Type 2 Infection-Induced Expression of CXCR3 Ligands Promotes CD4 + T Cell Migration and Is Regulated by the Viral Immediate-Early Protein ICP4. Front Immunol 2018; 9:2932. [PMID: 30619292 PMCID: PMC6305738 DOI: 10.3389/fimmu.2018.02932] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/29/2018] [Indexed: 12/18/2022] Open
Abstract
HSV-2 infection-induced CXCR3 ligands are important for the recruitment of virus-specific CD8+ T cells, but their impact on CD4+ T cell trafficking remains to be further determined. Given that recruitment of CD4+ T cells to infection areas may be one of the mechanisms that account for HSV-2 infection-mediated enhancement of HIV-1 sexual transmission, here we investigated the functionality of HSV-2 infection-induced CXCR3 ligands CXCL9, CXCL10, and CXCL11 in vivo and in vitro, and determined the viral components responsive for such induction and the underlying mechanisms. We first found that the expression of CXCR3 ligands CXCL9, CXCL10, and CXCL11 was increased in mice following vaginal challenge with HSV-2, while CXCL9 played a predominant role in the recruitment of CD4+ T cells to the vaginal foci of infected mice. HSV-2 infection also induced the production of CXCL9, CXCL10, and CXCL11 in human cervical epithelial cells. Of note, although HSV-2 induced the expression of all the three CXCR3 ligands, the induced CXCL9 appeared to play a predominant role in promoting CD4+ T cell migration, reflecting that the concentrations of CXCL10 and CXCL11 required for CD4+ T cell migration are higher than that of CXCL9. We further revealed that, ICP4, an immediate-early protein of HSV-2, is crucial in promoting CXCR3 ligand expression through the activation of p38 MAPK pathway. Mechanistically, ICP4 binds to corresponding promoters of CXCR3 ligands via interacting with the TATA binding protein (TBP), resulting in the transcriptional activation of the corresponding promoters. Taken together, our study highlights HSV-2 ICP4 as a vital viral protein in promoting CXCR3 ligand expression and CXCL9 as the key induced chemokine in mediating CD4+ T cell migration. Findings in this study have shed light on HSV-2 induced leukocyte recruitment which may be important for understanding HSV-2 infection-enhanced HIV-1 sexual transmission and the development of intervention strategies.
Collapse
Affiliation(s)
- Mudan Zhang
- The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Wuhan Institute of Virology, Chinese Academy of Science, Wuhan, China
| | - Xu Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xinmeng Guan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ming Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Binman Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Rui Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huimin Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yalan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Institute for Infection and Immunity, St George's University of London, London, United Kingdom
| |
Collapse
|
99921
|
Novel roles of Drosophila FUS and Aub responsible for piRNA biogenesis in neuronal disorders. Brain Res 2018; 1708:207-219. [PMID: 30578769 DOI: 10.1016/j.brainres.2018.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/16/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023]
Abstract
piRNAs, small non-coding RNAs, were considered to be restricted to germline cells. Although they have recently been detected in somatic cells including neurons, it remains unclear how piRNA biogenesis is involved in neuronal diseases. We herein examined the possible roles of Aubergine (Aub), a Piwi-family protein (PIWI) responsible for piRNA biogenesis, in the neuronal disorders, using the Cabeza (Caz) knockdown Drosophila. Caz is a Drosophila homologue of FUS, which is one of the genes causing amyotrophic lateral sclerosis (ALS). Aub overexpression enhanced the mobility defects accompanied by anatomical defects in motoneurons at neuromuscular junctions induced by the neuron-specific knockdown of Caz. In order to elucidate the underlying mechanisms, we examined pre-piRNA and mature-size piRNA levels under these conditions. qRT-PCR and RNA-seq analyses revealed that the Caz knockdown increased pre-piRNA levels, but reduced mature-size piRNA levels in the central nervous system (CNS), suggesting a role in the pre-piRNAs production. Aub overexpression did not increase mature-size piRNA levels. These results suggest that the accumulated pre-piRNAs are abnormal abortive pre-piRNAs that cannot be further processed by slicers, including Aub. We also demonstrated a relationship between Caz and pre-piRNAs in the CNS by RNA immunoprecipitation. Aub overexpression induced the abnormal cytoplasmic localization of Caz. Based on these results, we propose a model in which Caz knockdown-induced abnormal pre-piRNAs associate with Caz, then translocate and accumulate in the cytoplasm, a process that may be mediated by Aub. The novel roles for Caz and Aub demonstrated herein using the Caz-knockdown fly will contribute to a deeper understanding of the pathogenesis of ALS.
Collapse
|
99922
|
Kim Y, Jung JP, Pack CG, Huh WK. Global analysis of protein homomerization in Saccharomyces cerevisiae. Genome Res 2018; 29:135-145. [PMID: 30567710 PMCID: PMC6314163 DOI: 10.1101/gr.231860.117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/26/2018] [Indexed: 11/24/2022]
Abstract
In vivo analyses of the occurrence, subcellular localization, and dynamics of protein–protein interactions (PPIs) are important issues in functional proteomic studies. The bimolecular fluorescence complementation (BiFC) assay has many advantages in that it provides a reliable way to detect PPIs in living cells with minimal perturbation of the structure and function of the target proteins. Previously, to facilitate the application of the BiFC assay to genome-wide analysis of PPIs, we generated a collection of yeast strains expressing full-length proteins tagged with the N-terminal fragment of Venus (VN), a yellow fluorescent protein variant, from their own native promoters. In the present study, we constructed a VC (the C-terminal fragment of Venus) fusion library consisting of 5671 MATα strains expressing C-terminally VC-tagged proteins (representing ∼91% of the yeast proteome). For genome-wide analysis of protein homomer formation, we mated each strain in the VC fusion library with its cognate strain in the VN fusion library and performed the BiFC assay. From this analysis, we identified 186 homomer candidates. We further investigated the functional relevance of the homomerization of Pln1, a yeast perilipin. Our data set provides a useful resource for understanding the physiological roles of protein homomerization. Furthermore, the VC fusion library together with the VN fusion library will provide a valuable platform to systematically analyze PPIs in the natural cellular context.
Collapse
Affiliation(s)
- Yeonsoo Kim
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jong Pil Jung
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Chan-Gi Pack
- ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Won-Ki Huh
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Microbiology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
99923
|
Effect of Herring Antifreeze Protein Combined with Chitosan Magnetic Nanoparticles on Quality Attributes in Red Sea Bream (Pagrosomus major). FOOD BIOPROCESS TECH 2018. [DOI: 10.1007/s11947-018-2220-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
99924
|
Dysfunctional autophagy induced by the pro-apoptotic natural compound climacostol in tumour cells. Cell Death Dis 2018; 10:10. [PMID: 30584259 PMCID: PMC6315039 DOI: 10.1038/s41419-018-1254-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/06/2018] [Accepted: 12/03/2018] [Indexed: 01/22/2023]
Abstract
Autophagy occurs at a basal level in all eukaryotic cells and may support cell survival or activate death pathways. Due to its pathophysiologic significance, the autophagic machinery is a promising target for the development of multiple approaches for anti-neoplastic agents. We have recently described the cytotoxic and pro-apoptotic mechanisms, targeting the tumour suppressor p53, of climacostol, a natural product of the ciliated protozoan Climacostomum virens. We report here on how climacostol regulates autophagy and the involvement of p53-dependent mechanisms. Using both in vitro and in vivo techniques, we show that climacostol potently and selectively impairs autophagy in multiple tumour cells that are committed to die by apoptosis. In particular, in B16-F10 mouse melanomas climacostol exerts a marked and sustained accumulation of autophagosomes as the result of dysfunctional autophagic degradation. We also provide mechanistic insights showing that climacostol affects autophagosome turnover via p53-AMPK axis, although the mTOR pathway unrelated to p53 levels plays a role. In particular, climacostol activated p53 inducing the upregulation of p53 protein levels in the nuclei through effects on p53 stability at translational level, as for instance the phosphorylation at Ser15 site. Noteworthy, AMPKα activation was the major responsible of climacostol-induced autophagy disruption in the absence of a key role regulating cell death, thus indicating that climacostol effects on autophagy and apoptosis are two separate events, which may act independently on life/death decisions of the cell. Since the activation of p53 system is at the molecular crossroad regulating both the anti-autophagic action of climacostol and its role in the apoptosis induction, it might be important to explore the dual targeting of autophagy and apoptosis with agents acting on p53 for the selective killing of tumours. These findings also suggest the efficacy of ciliate bioactive molecules to identify novel lead compounds in drug discovery and development.
Collapse
|
99925
|
Niga P, Hansson-Mille PM, Swerin A, Claesson PM, Schoelkopf J, Gane PAC, Dai J, Furó I, Campbell RA, Johnson CM. Propofol adsorption at the air/water interface: a combined vibrational sum frequency spectroscopy, nuclear magnetic resonance and neutron reflectometry study. SOFT MATTER 2018; 15:38-46. [PMID: 30516226 DOI: 10.1039/c8sm01677a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Propofol is an amphiphilic small molecule that strongly influences the function of cell membranes, yet data regarding interfacial properties of propofol remain scarce. Here we consider propofol adsorption at the air/water interface as elucidated by means of vibrational sum frequency spectroscopy (VSFS), neutron reflectometry (NR), and surface tensiometry. VSFS data show that propofol adsorbed at the air/water interface interacts with water strongly in terms of hydrogen bonding and weakly in the proximity of the hydrocarbon parts of the molecule. In the concentration range studied there is almost no change in the orientation adopted at the interface. Data from NR show that propofol forms a dense monolayer with a thickness of 8.4 Å and a limiting area per molecule of 40 Å2, close to the value extracted from surface tensiometry. The possibility that islands or multilayers of propofol form at the air/water interface is therefore excluded as long as the solubility limit is not exceeded. Additionally, measurements of the 1H NMR chemical shifts demonstrate that propofol does not form dimers or multimers in bulk water up to the solubility limit.
Collapse
Affiliation(s)
- Petru Niga
- RISE Research Institutes of Sweden - Chemistry, Materials and Surfaces, Box 5607, SE-114 86 Stockholm, Sweden.
| | - Petra M Hansson-Mille
- RISE Research Institutes of Sweden - Chemistry, Materials and Surfaces, Box 5607, SE-114 86 Stockholm, Sweden.
| | - Agne Swerin
- RISE Research Institutes of Sweden - Chemistry, Materials and Surfaces, Box 5607, SE-114 86 Stockholm, Sweden. and KTH Royal Institute of Technology, Department of Chemistry, Division of Surface and Corrosion Science, SE-100 44 Stockholm, Sweden.
| | - Per M Claesson
- RISE Research Institutes of Sweden - Chemistry, Materials and Surfaces, Box 5607, SE-114 86 Stockholm, Sweden. and KTH Royal Institute of Technology, Department of Chemistry, Division of Surface and Corrosion Science, SE-100 44 Stockholm, Sweden.
| | | | - Patrick A C Gane
- Omya International AG, Baslerstrasse 42, CH-4665 Oftringen, Switzerland and Aalto University, School of Chemical Technology, Department of Bioproducts and Biosystems, FI-00076 Aalto, Helsinki, Finland
| | - Jing Dai
- KTH Royal Institute of Technology, Department of Chemistry, Division of Applied Physical Chemistry, SE-100 44 Stockholm, Sweden
| | - István Furó
- KTH Royal Institute of Technology, Department of Chemistry, Division of Applied Physical Chemistry, SE-100 44 Stockholm, Sweden
| | - Richard A Campbell
- Institut Laue-Langevin, 71 Avenue des Martyrs, CS20156, 38042 Grenoble Cedex 9, France and Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK
| | - C Magnus Johnson
- KTH Royal Institute of Technology, Department of Chemistry, Division of Surface and Corrosion Science, SE-100 44 Stockholm, Sweden.
| |
Collapse
|
99926
|
Grafanaki K, Anastasakis D, Kyriakopoulos G, Skeparnias I, Georgiou S, Stathopoulos C. Translation regulation in skin cancer from a tRNA point of view. Epigenomics 2018; 11:215-245. [PMID: 30565492 DOI: 10.2217/epi-2018-0176] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Protein synthesis is a central and dynamic process, frequently deregulated in cancer through aberrant activation or expression of translation initiation factors and tRNAs. The discovery of tRNA-derived fragments, a new class of abundant and, in some cases stress-induced, small Noncoding RNAs has perplexed the epigenomics landscape and highlights the emerging regulatory role of tRNAs in translation and beyond. Skin is the biggest organ in human body, which maintains homeostasis of its multilayers through regulatory networks that induce translational reprogramming, and modulate tRNA transcription, modification and fragmentation, in response to various stress signals, like UV irradiation. In this review, we summarize recent knowledge on the role of translation regulation and tRNA biology in the alarming prevalence of skin cancer.
Collapse
Affiliation(s)
- Katerina Grafanaki
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece.,Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Dimitrios Anastasakis
- National Institute of Musculoskeletal & Arthritis & Skin, NIH, 50 South Drive, Room 1152, Bethesda, MD 20892, USA
| | - George Kyriakopoulos
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Ilias Skeparnias
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Sophia Georgiou
- Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | | |
Collapse
|
99927
|
Poly(ADP-Ribose) Polymerases in Host-Pathogen Interactions, Inflammation, and Immunity. Microbiol Mol Biol Rev 2018; 83:83/1/e00038-18. [PMID: 30567936 DOI: 10.1128/mmbr.00038-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The literature review presented here details recent research involving members of the poly(ADP-ribose) polymerase (PARP) family of proteins. Among the 17 recognized members of the family, the human enzyme PARP1 is the most extensively studied, resulting in a number of known biological and metabolic roles. This review is focused on the roles played by PARP enzymes in host-pathogen interactions and in diseases with an associated inflammatory response. In mammalian cells, several PARPs have specific roles in the antiviral response; this is perhaps best illustrated by PARP13, also termed the zinc finger antiviral protein (ZAP). Plant stress responses and immunity are also regulated by poly(ADP-ribosyl)ation. PARPs promote inflammatory responses by stimulating proinflammatory signal transduction pathways that lead to the expression of cytokines and cell adhesion molecules. Hence, PARP inhibitors show promise in the treatment of inflammatory disorders and conditions with an inflammatory component, such as diabetes, arthritis, and stroke. These functions are correlated with the biophysical characteristics of PARP family enzymes. This work is important in providing a comprehensive understanding of the molecular basis of pathogenesis and host responses, as well as in the identification of inhibitors. This is important because the identification of inhibitors has been shown to be effective in arresting the progression of disease.
Collapse
|
99928
|
Ashraf U, Benoit-Pilven C, Lacroix V, Navratil V, Naffakh N. Advances in Analyzing Virus-Induced Alterations of Host Cell Splicing. Trends Microbiol 2018; 27:268-281. [PMID: 30577974 DOI: 10.1016/j.tim.2018.11.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/19/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022]
Abstract
Alteration of host cell splicing is a common feature of many viral infections which is underappreciated because of the complexity and technical difficulty of studying alternative splicing (AS) regulation. Recent advances in RNA sequencing technologies revealed that up to several hundreds of host genes can show altered mRNA splicing upon viral infection. The observed changes in AS events can be either a direct consequence of viral manipulation of the host splicing machinery or result indirectly from the virus-induced innate immune response or cellular damage. Analysis at a higher resolution with single-cell RNAseq, and at a higher scale with the integration of multiple omics data sets in a systems biology perspective, will be needed to further comprehend this complex facet of virus-host interactions.
Collapse
Affiliation(s)
- Usama Ashraf
- Institut Pasteur, Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, F-75015 Paris, France; CNRS UMR3569, F-75015 Paris, France; Université Paris Diderot, Sorbonne Paris Cité EA302, F-75015 Paris, France
| | - Clara Benoit-Pilven
- INSERM U1028; CNRS UMR5292, Lyon Neuroscience Research Center, Genetic of Neuro-development Anomalies Team, F-69000 Lyon, France; Université Claude Bernard Lyon 1, CNRS UMR5558, Laboratoire de Biométrie et Biologie Evolutive, F-69622 Villeurbanne, France; EPI ERABLE, INRIA Grenoble Rhône-Alpes, F-38330 Montbonnot Saint-Martin, France
| | - Vincent Lacroix
- Université Claude Bernard Lyon 1, CNRS UMR5558, Laboratoire de Biométrie et Biologie Evolutive, F-69622 Villeurbanne, France; EPI ERABLE, INRIA Grenoble Rhône-Alpes, F-38330 Montbonnot Saint-Martin, France
| | - Vincent Navratil
- PRABI, Rhône Alpes Bioinformatics Center, UCBL, Université Claude Bernard Lyon 1, F-69000 Lyon, France; European Virus Bioinformatics Center, Leutragraben 1, D-07743 Jena, Germany
| | - Nadia Naffakh
- Institut Pasteur, Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, F-75015 Paris, France; CNRS UMR3569, F-75015 Paris, France; Université Paris Diderot, Sorbonne Paris Cité EA302, F-75015 Paris, France.
| |
Collapse
|
99929
|
Morris RJ. Thy-1, a Pathfinder Protein for the Post-genomic Era. Front Cell Dev Biol 2018; 6:173. [PMID: 30619853 PMCID: PMC6305390 DOI: 10.3389/fcell.2018.00173] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Thy-1 is possibly the smallest of cell surface proteins – 110 amino acids folded into an Immunoglobulin variable domain, tethered to the outer leaflet of the cell surface membrane via just the two saturated fatty acids of its glycosylphosphatidylinositol (GPI) anchor. Yet Thy-1 is emerging as a key regulator of differentiation in cells of endodermal, mesodermal, and ectodermal origin, acting as both a ligand (for certain integrins and other receptors), and as a receptor, able to modulate signaling and hence differentiation in the Thy-1-expressing cell. This is an extraordinary diversity of molecular pathways to be controlled by a molecule that does not even cross the cell membrane. Here I review aspects of the cell biology of Thy-1, and studies of its role as deduced from gene knock-out studies, that suggest how this protein can participate in so many different signaling-related functions. While mechanisms differ in molecular detail, it appears overall that Thy-1 dampens down signaling to control function.
Collapse
Affiliation(s)
- Roger J Morris
- Department of Chemistry, King's College London, London, United Kingdom
| |
Collapse
|
99930
|
Jhun BS, O-Uchi J, Adaniya SM, Cypress MW, Yoon Y. Adrenergic Regulation of Drp1-Driven Mitochondrial Fission in Cardiac Physio-Pathology. Antioxidants (Basel) 2018; 7:antiox7120195. [PMID: 30567380 PMCID: PMC6316402 DOI: 10.3390/antiox7120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/28/2022] Open
Abstract
Abnormal mitochondrial morphology, especially fragmented mitochondria, and mitochondrial dysfunction are hallmarks of a variety of human diseases including heart failure (HF). Although emerging evidence suggests a link between mitochondrial fragmentation and cardiac dysfunction, it is still not well described which cardiac signaling pathway regulates mitochondrial morphology and function under pathophysiological conditions such as HF. Mitochondria change their shape and location via the activity of mitochondrial fission and fusion proteins. This mechanism is suggested as an important modulator for mitochondrial and cellular functions including bioenergetics, reactive oxygen species (ROS) generation, spatiotemporal dynamics of Ca2+ signaling, cell growth, and death in the mammalian cell- and tissue-specific manners. Recent reports show that a mitochondrial fission protein, dynamin-like/related protein 1 (DLP1/Drp1), is post-translationally modified via cell signaling pathways, which control its subcellular localization, stability, and activity in cardiomyocytes/heart. In this review, we summarize the possible molecular mechanisms for causing post-translational modifications (PTMs) of DLP1/Drp1 in cardiomyocytes, and further discuss how these PTMs of DLP1/Drp1 mediate abnormal mitochondrial morphology and mitochondrial dysfunction under adrenergic signaling activation that contributes to the development and progression of HF.
Collapse
Affiliation(s)
- Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA.
- Department of Medicine, Division of Cardiology, the Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
99931
|
Abstract
In cells, intra- and intermolecular interactions of proteins confer function, and the dynamic modulation of this interactome is critical to meet the changing needs required to support life. Cross-linking and mass spectrometry (XL-MS) enable the detection of both intra- and intermolecular protein interactions in organelles, cells, tissues, and organs. Quantitative XL-MS enables the detection of interactome changes in cells due to environmental, phenotypic, pharmacological, or genetic perturbations. We have developed new informatics capabilities, the first to enable 3D visualization of multiple quantitative interactome data sets, acquired over time or with varied perturbation levels, to reveal relevant dynamic interactome changes. These new tools are integrated within release 3.0 of our online cross-linked peptide database and analysis tool suite XLinkDB. With the recent rapid expansion in XL-MS for protein structural studies and the extension to quantitative XL-MS measurements, 3D interactome visualization tools are of critical need.
Collapse
Affiliation(s)
- Andrew Keller
- Department of Genome Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Juan D Chavez
- Department of Genome Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Jimmy K Eng
- Department of Genome Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - Zorian Thornton
- Department of Genome Sciences , University of Washington , Seattle , Washington 98195 , United States
| | - James E Bruce
- Department of Genome Sciences , University of Washington , Seattle , Washington 98195 , United States
| |
Collapse
|
99932
|
Yang B, Adams DJ, Marlow M, Zelzer M. Surface-Mediated Supramolecular Self-Assembly of Protein, Peptide, and Nucleoside Derivatives: From Surface Design to the Underlying Mechanism and Tailored Functions. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:15109-15125. [PMID: 30032622 DOI: 10.1021/acs.langmuir.8b01165] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Among the many parameters that have been explored to exercise control over self-assembly processes, the influence of surface properties on self-assembly has been recognized as important but has received considerably less attention than other factors. This is particularly true for biomolecule-derived self-assembling molecules such as protein, peptide, and nucleobase derivatives. Because of their relevance to biomaterial and drug delivery applications, interest in these materials is increasing. As the formation of supramolecular structures from these biomolecule derivatives inevitably brings them into contact with the surfaces of surrounding materials, understanding and controlling the impact of the properties of these surfaces on the self-assembly process are important. In this feature article, we present an overview of the different surface parameters that have been used and studied for the direction of the self-assembly of protein, peptide, and nucleoside-based molecules. The current mechanistic understanding of these processes will be discussed, and potential applications of surface-mediated self-assembly will be outlined.
Collapse
Affiliation(s)
- Bin Yang
- Department of Pharmacy , University of Nottingham , Nottingham NG2 7RD , U.K
| | - Dave J Adams
- School of Chemistry , University of Glasgow , Glasgow G12 8QQ , U.K
| | - Maria Marlow
- Department of Pharmacy , University of Nottingham , Nottingham NG2 7RD , U.K
| | - Mischa Zelzer
- Department of Pharmacy , University of Nottingham , Nottingham NG2 7RD , U.K
| |
Collapse
|
99933
|
Houwman JA, Westphal AH, Visser AJWG, Borst JW, van Mierlo CPM. Concurrent presence of on- and off-pathway folding intermediates of apoflavodoxin at physiological ionic strength. Phys Chem Chem Phys 2018; 20:7059-7072. [PMID: 29473921 DOI: 10.1039/c7cp07922b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Flavodoxins have a protein topology that can be traced back to the universal ancestor of the three kingdoms of life. Proteins with this type of architecture tend to temporarily misfold during unassisted folding to their native state and form intermediates. Several of these intermediate species are molten globules (MGs), which are characterized by a substantial amount of secondary structure, yet without the tertiary side-chain packing of natively folded proteins. An off-pathway MG is formed at physiological ionic strength in the case of the F44Y variant of Azotobacter vinelandii apoflavodoxin (i.e., flavodoxin without flavin mononucleotide (FMN)). Here, we show that at this condition actually two folding species of this apoprotein co-exist at equilibrium. These species were detected by using a combination of FMN fluorescence quenching upon cofactor binding to the apoprotein and of polarized time-resolved tryptophan fluorescence spectroscopy. Besides the off-pathway MG, we observe the simultaneous presence of an on-pathway folding intermediate, which is native-like. Presence of concurrent intermediates at physiological ionic strength enables future exploration of how aspects of the cellular environment, like for example involvement of chaperones, affect these species.
Collapse
Affiliation(s)
- Joseline A Houwman
- Laboratory of Biochemistry, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| | | | | | | | | |
Collapse
|
99934
|
Ohui K, Afanasenko E, Bacher F, Ting RLX, Zafar A, Blanco-Cabra N, Torrents E, Dömötör O, May NV, Darvasiova D, Enyedy ÉA, Popović-Bijelić A, Reynisson J, Rapta P, Babak MV, Pastorin G, Arion VB. New Water-Soluble Copper(II) Complexes with Morpholine-Thiosemicarbazone Hybrids: Insights into the Anticancer and Antibacterial Mode of Action. J Med Chem 2018; 62:512-530. [PMID: 30507173 PMCID: PMC6348444 DOI: 10.1021/acs.jmedchem.8b01031] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Six
morpholine-(iso)thiosemicarbazone hybrids HL1–HL6 and
their Cu(II) complexes with good-to-moderate solubility and
stability in water were synthesized and characterized. Cu(II) complexes [Cu(L1–6)Cl] (1–6) formed weak dimeric associates in the solid state,
which did not remain intact in solution as evidenced by ESI-MS. The
lead proligands and Cu(II) complexes displayed higher antiproliferative
activity in cancer cells than triapine. In addition, complexes 2–5 were found to specifically inhibit the growth of
Gram-positive bacteria Staphylococcus aureus with MIC50 values at 2–5 μg/mL. Insights
into the processes controlling intracellular accumulation and mechanism
of action were investigated for 2 and 5,
including the role of ribonucleotide reductase (RNR) inhibition, endoplasmic
reticulum stress induction, and regulation of other cancer signaling
pathways. Their ability to moderately inhibit R2 RNR protein in the
presence of dithiothreitol is likely related to Fe chelating properties
of the proligands liberated upon reduction.
Collapse
Affiliation(s)
- Kateryna Ohui
- Institute of Inorganic Chemistry , University of Vienna , Währinger Strasse 42 , A-1090 Vienna , Austria
| | - Eleonora Afanasenko
- Institute of Inorganic Chemistry , University of Vienna , Währinger Strasse 42 , A-1090 Vienna , Austria
| | - Felix Bacher
- Institute of Inorganic Chemistry , University of Vienna , Währinger Strasse 42 , A-1090 Vienna , Austria
| | - Rachel Lim Xue Ting
- Department of Pharmacy , National University of Singapore , 3 Science Drive 2 , Singapore 117543 , Singapore
| | - Ayesha Zafar
- School of Chemical Sciences , University of Auckland , Auckland 1010 , New Zealand
| | - Núria Blanco-Cabra
- Bacterial Infections: Antimicrobial Therapies, Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Barcelona 08036 , Spain
| | - Eduard Torrents
- Bacterial Infections: Antimicrobial Therapies, Institute for Bioengineering of Catalonia (IBEC) , The Barcelona Institute of Science and Technology , Barcelona 08036 , Spain
| | - Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7. , H-6720 Szeged , Hungary
| | - Nóra V May
- Research Centre of Natural Sciences , Hungarian Academy of Sciences , Magyar tudósok körútja 2. , H-1117 Budapest , Hungary
| | - Denisa Darvasiova
- Institute of Physical Chemistry and Chemical Physics , Slovak Technical University of Technology , Radlinského 9 , 81237 Bratislava , Slovak Republic
| | - Éva A Enyedy
- Department of Inorganic and Analytical Chemistry , University of Szeged , Dóm tér 7. , H-6720 Szeged , Hungary
| | - Ana Popović-Bijelić
- Faculty of Physical Chemistry , University of Belgrade , 11158 Belgrade , Serbia
| | - Jóhannes Reynisson
- School of Chemical Sciences , University of Auckland , Auckland 1010 , New Zealand
| | - Peter Rapta
- Institute of Physical Chemistry and Chemical Physics , Slovak Technical University of Technology , Radlinského 9 , 81237 Bratislava , Slovak Republic
| | - Maria V Babak
- Department of Chemistry , National University of Singapore , 3 Science Drive 2 , 117543 , Singapore.,Drug Development Unit , National University of Singapore , 28 Medical Drive , 117546 , Singapore
| | - Giorgia Pastorin
- Department of Pharmacy , National University of Singapore , 3 Science Drive 2 , Singapore 117543 , Singapore
| | - Vladimir B Arion
- Institute of Inorganic Chemistry , University of Vienna , Währinger Strasse 42 , A-1090 Vienna , Austria
| |
Collapse
|
99935
|
Metabolic Signaling into Chromatin Modifications in the Regulation of Gene Expression. Int J Mol Sci 2018; 19:ijms19124108. [PMID: 30567372 PMCID: PMC6321258 DOI: 10.3390/ijms19124108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
Abstract
The regulation of cellular metabolism is coordinated through a tissue cross-talk by hormonal control. This leads to the establishment of specific transcriptional gene programs which adapt to environmental stimuli. On the other hand, recent advances suggest that metabolic pathways could directly signal into chromatin modifications and impact on specific gene programs. The key metabolites acetyl-CoA or S-adenosyl-methionine (SAM) are examples of important metabolic hubs which play in addition a role in chromatin acetylation and methylation. In this review, we will discuss how intermediary metabolism impacts on transcription regulation and the epigenome with a particular focus in metabolic disorders.
Collapse
|
99936
|
Hoffmann M, Pioch M, Pralow A, Hennig R, Kottler R, Reichl U, Rapp E. The Fine Art of Destruction: A Guide to In-Depth Glycoproteomic Analyses-Exploiting the Diagnostic Potential of Fragment Ions. Proteomics 2018; 18:e1800282. [DOI: 10.1002/pmic.201800282] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/07/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Marcus Hoffmann
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
| | - Markus Pioch
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
| | - Alexander Pralow
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
| | - René Hennig
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
- glyXera GmbH; 39120 Magdeburg Germany
| | - Robert Kottler
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
- glyXera GmbH; 39120 Magdeburg Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
- Chair of Bioprocess Engineering; Otto von Guericke University Magdeburg; 39106 Magdeburg Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems; Bioprocess Engineering; 39106 Magdeburg Germany
- glyXera GmbH; 39120 Magdeburg Germany
| |
Collapse
|
99937
|
Physiological and TMT-based proteomic analysis of oat early seedlings in response to alkali stress. J Proteomics 2018; 193:10-26. [PMID: 30576833 DOI: 10.1016/j.jprot.2018.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/07/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Oats are an important cereal crop worldwide, and they also serve as a phytoremediation crop to ameliorate salinized and alkalized soils. However, the mechanism of the oat response to alkali remains unclear. Physiological and tandem mass tag (TMT)-based proteomic analyses were employed to elucidate the mechanism of the oat response to alkali stress. Physiological and phenotypic data showed that oat root growth was inhibited more severely than shoot growth after alkali stress. In total, 164 proteins were up-regulated and 241 proteins were down-regulated in roots, and 93 proteins were up-regulated and 139 proteins were down-regulated in shoots. Under high pH stress, transmembrane proton transporters were down-regulated; conversely, organic acid synthesis related enzymes were increased. Transporters of N, P, Fe, Cu and Ca in addition to N assimilation enzymes in the root were highly increased. This result revealed that higher efficiency of P, Fe, Cu and Ca transport, especially higher efficiency of N intake and assimilation, greatly promoted oat root resistance to alkali stress. Furthermore, many resistance proteins, such as late embryogenesis abundant (LEA) mainly in shoots, GDSL esterase lipase mainly in roots, and WD40-like beta propeller repeat families, greatly accumulated to contribute to oat resistance to alkali stress. SIGNIFICANCE: In this study, physiological and tandem mass tag (TMT)-based proteomic analyses were employed to elucidate oats early seedlings in response to alkali stress. Many difference expression proteins were found involving in oats response to alkali stress. Also, higher efficiency transport of P, Fe, Cu, Ca and N greatly promoted oat resistance to alkali stress.
Collapse
|
99938
|
Pan Y, Zagorski K, Shlyakhtenko LS, Lyubchenko YL. The Enzymatic Activity of APOBE3G Multimers. Sci Rep 2018; 8:17953. [PMID: 30560880 PMCID: PMC6298963 DOI: 10.1038/s41598-018-36372-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/02/2018] [Indexed: 11/19/2022] Open
Abstract
APOBEC3G (A3G) belongs to the family of cytosine deaminases that play an important role in the innate immune response. Similar to other, two-domain members of the APOBEC family, A3G is prone to concentration-dependent oligomerization, which is an integral for its function in the cell. It is shown that oligomerization of A3G is related to the packing mechanism into virus particle and, is critical for the so-called roadblock model during reverse transcription of proviral ssDNA. The role of oligomerization for deaminase activity of A3G is widely discussed in the literature; however, its relevance to deaminase activity for different oligomeric forms of A3G remains unclear. Here, using Atomic Force Microscopy, we directly visualized A3G-ssDNA complexes, determined their yield and stoichiometry and in parallel, using PCR assay, measured the deaminase activity of these complexes. Our data demonstrate a direct correlation between the total yield of A3G-ssDNA complexes and their total deaminase activity. Using these data, we calculated the relative deaminase activity for each individual oligomeric state of A3G in the complex. Our results show not only similar deaminase activity for monomer, dimer and tetramer of A3G in the complex, but indicate that larger oligomers of A3G retain their deaminase activity.
Collapse
Affiliation(s)
- Yangang Pan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, 68198-6025, USA
| | - Karen Zagorski
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, 68198-6025, USA
| | - Luda S Shlyakhtenko
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, 68198-6025, USA.
| | - Yuri L Lyubchenko
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, 68198-6025, USA.
| |
Collapse
|
99939
|
Shimizu K, Matsuoka Y. Regulation of glycolytic flux and overflow metabolism depending on the source of energy generation for energy demand. Biotechnol Adv 2018; 37:284-305. [PMID: 30576718 DOI: 10.1016/j.biotechadv.2018.12.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/06/2018] [Accepted: 12/15/2018] [Indexed: 12/11/2022]
Abstract
Overflow metabolism is a common phenomenon observed at higher glycolytic flux in many bacteria, yeast (known as Crabtree effect), and mammalian cells including cancer cells (known as Warburg effect). This phenomenon has recently been characterized as the trade-offs between protein costs and enzyme efficiencies based on coarse-graining approaches. Moreover, it has been recognized that the glycolytic flux increases as the source of energy generation changes from energetically efficient respiration to inefficient respiro-fermentative or fermentative metabolism causing overflow metabolism. It is highly desired to clarify the metabolic regulation mechanisms behind such phenomena. Metabolic fluxes are located on top of the hierarchical regulation systems, and represent the outcome of the integrated response of all levels of cellular regulation systems. In the present article, we discuss about the different levels of regulation systems for the modulation of fluxes depending on the growth rate, growth condition such as oxygen limitation that alters the metabolism towards fermentation, and genetic perturbation affecting the source of energy generation from respiration to respiro-fermentative metabolism in relation to overflow metabolism. The intracellular metabolite of the upper glycolysis such as fructose 1,6-bisphosphate (FBP) plays an important role not only for flux sensing, but also for the regulation of the respiratory activity either directly or indirectly (via transcription factors) at higher growth rate. The glycolytic flux regulation is backed up (enhanced) by unphosphorylated EIIA and HPr of the phosphotransferase system (PTS) components, together with the sugar-phosphate stress regulation, where the transcriptional regulation is further modulated by post-transcriptional regulation via the degradation of mRNA (stability of mRNA) in Escherichia coli. Moreover, the channeling may also play some role in modulating the glycolytic cascade reactions.
Collapse
Affiliation(s)
- Kazuyuki Shimizu
- Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan; Institute of Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan.
| | - Yu Matsuoka
- Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| |
Collapse
|
99940
|
Ascenzi P, De Simone G, Ciaccio C, Santucci R, Coletta M. Hydroxylamine-induced oxidation of ferrous CO-bound carboxymethylated-cytochrome c. J PORPHYR PHTHALOCYA 2018. [DOI: 10.1142/s1088424618501055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The hexa-coordinated metal center of horse heart cyt[Formula: see text] (cyt[Formula: see text] is at the root of its low reactivity. In contrast, carboxymethylated cyt[Formula: see text] (CM-cyt[Formula: see text] displays myoglobin-like properties. Herein, kinetics of CO binding to ferrous CM-cyt[Formula: see text] (CM-cyt[Formula: see text](II)) and of the irreversible oxidation of ferrous carbonylated CM-cyt[Formula: see text] (CM-cyt[Formula: see text](II)-CO) by hydroxylamine (HA), at pH 5.8 and 20.0 [Formula: see text]C, are reported. HA irreversibly oxidizes CM-cyt[Formula: see text](II)-CO with the 1:2 stoichiometry leading to the formation of the ferric species (CM-cyt[Formula: see text](III)) without the observation of intermediates. Present data indicate that: (i) the rate of CO dissociation from CM-cyt[Formula: see text](II)-CO represents the rate-limiting step of HA-mediated oxidation of the carbonylated metal center, (ii) the fast oxidation of CM-cyt[Formula: see text](II)-CO from HA reflects the penta-coordination of the transient CM-cyt[Formula: see text](II) species, (iii) the HA-catalyzed conversion of CM-cyt[Formula: see text](II)-CO to CM-cyt[Formula: see text](III) could proceed via the geminate mechanism, (iv) values of the second-order rate constants for the carbonylation and the HA-mediated oxidation of ferrous heme-proteins are linearly correlated reflecting the penta- or hexa-coordination of the metal center, the free energy for the in-plane positioning of the heme-Fe atom in the unliganded species, and the arrangement of the distal portion of the heme pocket that affects ligand and/or electron transfer.
Collapse
Affiliation(s)
- Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, I-00146 Roma, Italy
| | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma “Tor Vergata”, I-00133 Roma, Italy
- Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, I-70126 Bari, Italy
| | - Roberto Santucci
- Department of Clinical Sciences and Translational Medicine, University of Roma “Tor Vergata”, I-00133 Roma, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma “Tor Vergata”, I-00133 Roma, Italy
- Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, I-70126 Bari, Italy
| |
Collapse
|
99941
|
Abstract
The movement or trafficking of heme is critical for cellular functions (e.g., oxygen transport and energy production); however, intracellular heme is tightly regulated due to its inherent cytotoxicity. These factors, combined with the transient nature of transport, have resulted in a lack of direct knowledge on the mechanisms of heme binding and trafficking. Here, we used the cytochrome c biogenesis system II pathway as a model to study heme trafficking. System II is composed of two integral membrane proteins (CcsBA) which function to transport heme across the membrane and stereospecifically position it for covalent attachment to apocytochrome c. We mapped two heme binding domains in CcsBA and suggest a path for heme trafficking. These data, in combination with metagenomic coevolution data, are used to determine a structural model of CcsBA, leading to increased understanding of the mechanisms for heme transport and the cytochrome c synthetase function of CcsBA. Although intracellular heme trafficking must occur for heme protein assembly, only a few heme transporters have been unequivocally discovered and nothing is known about their structure or mechanisms. Cytochrome c biogenesis in prokaryotes requires the transport of heme from inside to outside for stereospecific attachment to cytochrome c via two thioether bonds (at CXXCH). The CcsBA integral membrane protein was shown to transport and attach heme (and thus is a cytochrome c synthetase), but the structure and mechanisms underlying these two activities are poorly understood. We employed a new cysteine/heme crosslinking tool that traps endogenous heme in heme binding sites. We combined these data with a comprehensive imidazole correction approach (for heme ligand interrogation) to map heme binding sites. Results illuminate the process of heme transfer through the membrane to an external binding site (called the WWD domain). Using meta-genomic data (GREMLIN) and Rosetta modeling programs, a structural model of the transmembrane (TM) regions in CcsBA were determined. The heme mapping data were then incorporated to model the TM heme binding site (with TM-His1 and TM-His2 as ligands) and the external heme binding WWD domain (with P-His1 and P-His2 as ligands). Other periplasmic structure/function studies facilitated modeling of the full CcsBA protein as a framework for understanding the mechanisms. Mechanisms are proposed for heme transport from TM-His to WWD/P-His and subsequent stereospecific attachment of heme. A ligand exchange of the P-His1 for histidine of CXXCH at the synthetase active site is suggested.
Collapse
|
99942
|
Yasir M, Icke C, Abdelwahab R, Haycocks JR, Godfrey RE, Sazinas P, Pallen MJ, Henderson IR, Busby SJW, Browning DF. Organization and architecture of AggR-dependent promoters from enteroaggregative Escherichia coli. Mol Microbiol 2018; 111:534-551. [PMID: 30485564 PMCID: PMC6392122 DOI: 10.1111/mmi.14172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2018] [Indexed: 11/27/2022]
Abstract
Enteroaggregative Escherichia coli (EAEC), is a diarrhoeagenic human pathogen commonly isolated from patients in both developing and industrialized countries. Pathogenic EAEC strains possess many virulence determinants, which are thought to be involved in causing disease, though, the exact mechanism by which EAEC causes diarrhoea is unclear. Typical EAEC strains possess the transcriptional regulator, AggR, which controls the expression of many virulence determinants, including the attachment adherence fimbriae (AAF) that are necessary for adherence to human gut epithelial cells. Here, using RNA‐sequencing, we have investigated the AggR regulon from EAEC strain 042 and show that AggR regulates the transcription of genes on both the bacterial chromosome and the large virulence plasmid, pAA2. Due to the importance of fimbriae, we focused on the two AAF/II fimbrial gene clusters in EAEC 042 (afaB‐aafCB and aafDA) and identified the promoter elements and AggR‐binding sites required for fimbrial expression. In addition, we examined the organization of the fimbrial operon promoters from other important EAEC strains to understand the rules of AggR‐dependent activation. Finally, we generated a series of semi‐synthetic promoters to define the minimal sequence required for AggR‐mediated activation and show that the correct positioning of a single AggR‐binding site is sufficient to confer AggR‐dependence.
Collapse
Affiliation(s)
- Muhammad Yasir
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.,Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Christopher Icke
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Radwa Abdelwahab
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.,Faculty of Medicine, Assiut University, Assiut, Egypt
| | - James R Haycocks
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Rita E Godfrey
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Pavelas Sazinas
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs Lyngby, Denmark
| | - Mark J Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Ian R Henderson
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Stephen J W Busby
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
99943
|
Firman T, Amgalan A, Ghosh K. Maximum Caliber Can Build and Infer Models of Oscillation in a Three-Gene Feedback Network. J Phys Chem A 2018. [DOI: 10.1021/acs.jpca.8b07465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
99944
|
A viral-fusion-peptide-like molecular switch drives membrane insertion of botulinum neurotoxin A1. Nat Commun 2018; 9:5367. [PMID: 30560862 PMCID: PMC6299077 DOI: 10.1038/s41467-018-07789-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 11/22/2018] [Indexed: 11/09/2022] Open
Abstract
Botulinum neurotoxin (BoNT) delivers its protease domain across the vesicle membrane to enter the neuronal cytosol upon vesicle acidification. This process is mediated by its translocation domain (HN), but the molecular mechanism underlying membrane insertion of HN remains poorly understood. Here, we report two crystal structures of BoNT/A1 HN that reveal a novel molecular switch (termed BoNT-switch) in HN, where buried α-helices transform into surface-exposed hydrophobic β-hairpins triggered by acidic pH. Locking the BoNT-switch by disulfide trapping inhibited the association of HN with anionic liposomes, blocked channel formation by HN, and reduced the neurotoxicity of BoNT/A1 by up to ~180-fold. Single particle counting studies showed that an acidic environment tends to promote BoNT/A1 self-association on liposomes, which is partly regulated by the BoNT-switch. These findings suggest that the BoNT-switch flips out upon exposure to the acidic endosomal pH, which enables membrane insertion of HN that subsequently leads to LC delivery. The translocation domain (HN) of Botulinum neurotoxins (BoNTs) mediates the delivery of the BoNT light chain (LC) into neuronal cytosol. Here the authors provide insights into HN membrane insertion by determining the crystal structure of BoNT/A1 HN at acidic pH, which reveals a molecular switch in HN, where buried α-helices are transformed into surface-exposed hydrophobic β-hairpins.
Collapse
|
99945
|
Rodriguez J, Zhang Y, Li T, Xie C, Sun Y, Xu Y, Zhou K, Huo K, Wang Y, Wang X, Andersson D, Ståhlberg A, Xing Q, Mallard C, Hagberg H, Modjtahedi N, Kroemer G, Blomgren K, Zhu C. Lack of the brain-specific isoform of apoptosis-inducing factor aggravates cerebral damage in a model of neonatal hypoxia-ischemia. Cell Death Dis 2018; 10:3. [PMID: 30584234 PMCID: PMC6315035 DOI: 10.1038/s41419-018-1250-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/15/2018] [Accepted: 12/03/2018] [Indexed: 12/22/2022]
Abstract
Apoptosis-inducing factor (AIF) may contribute to neuronal cell death, and its influence is particularly prominent in the immature brain after hypoxia-ischemia (HI). A brain-specific AIF splice-isoform (AIF2) has recently been discovered, but has not yet been characterized at the genetic level. The aim of this study was to determine the functional and regulatory profile of AIF2 under physiological conditions and after HI in mice. We generated AIF2 knockout (KO) mice by removing the AIF2-specific exon and found that the relative expression of Aif1 mRNA increased in Aif2 KO mice and that this increase became even more pronounced as Aif2 KO mice aged compared to their wild-type (WT) littermates. Mitochondrial morphology and function, reproductive function, and behavior showed no differences between WT and Aif2 KO mice. However, lack of AIF2 enhanced brain injury in neonatal mice after HI compared to WT controls, and this effect was linked to increased oxidative stress but not to caspase-dependent or -independent apoptosis pathways. These results indicate that AIF2 deficiency exacerbates free radical production and HI-induced neonatal brain injury.
Collapse
Affiliation(s)
- Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Yaodong Zhang
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Department of Pediatrics, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Li
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Department of Pediatrics, Children's Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Yanyan Sun
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yiran Xu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kai Zhou
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Kaiming Huo
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Pediatrics, Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yafeng Wang
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Department of Pediatrics, Children's Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Daniel Andersson
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Qinghe Xing
- Institute of Biomedical Science of Fudan University, Shanghai, 201102, China
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Henrik Hagberg
- Center for Perinatal Medicine and Health, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Guido Kroemer
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Metabolomics and Cell Biology Platforms, GRCC, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, INSERM U1138, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Labex Immuno-Oncology, Paris, France
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden.
- Henan Key Laboratory of Child Brain Injury, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
99946
|
Abstract
Function is an onerous concept, as the recent study by Steven Salzberg and colleagues demonstrates. We should be careful and always specific in using the ‘F-word’.
Collapse
Affiliation(s)
- W Ford Doolittle
- Department of Biochemistry and Molecular Biology, Dalhousie University Faculty of Medicine, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
99947
|
When safeguarding goes wrong: Impact of oxidative stress on protein homeostasis in health and neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:221-264. [PMID: 30635082 DOI: 10.1016/bs.apcsb.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular redox status is an established player in many different cellular functions. The buildup of oxidants within the cell is tightly regulated to maintain a balance between the positive and negative outcomes of cellular oxidants. Proteins are highly sensitive to oxidation, since modification can cause widespread unfolding and the formation of toxic aggregates. In response, cells have developed highly regulated systems that contribute to the maintenance of both the global redox status and protein homeostasis at large. Changes to these systems have been found to correlate with aging and age-related disorders, such as neurodegenerative pathologies. This raises intriguing questions as to the source of the imbalance in the redox and protein homeostasis systems, their interconnectivity, and their role in disease progression. Here we focus on the crosstalk between the redox and protein homeostasis systems in neurodegenerative diseases, specifically in Alzheimer's, Parkinson's, and ALS. We elaborate on some of the main players of the stress response systems, including the master regulators of oxidative stress and the heat shock response, Nrf2 and Hsf1, which are essential features of protein folding, and mediators of protein turnover. We illustrate the elegant mechanisms used by these components to provide an immediate response, including protein plasticity controlled by redox-sensing cysteines and the recruitment of naive proteins to the redox homeostasis array that act as chaperons in an ATP-independent manner.
Collapse
|
99948
|
Abstract
Francisella tularensis is a highly infectious intracellular pathogen that kills more than half of infected humans if left untreated. F. tularensis has also been classified as a potential bioterrorism agent with a great risk for deliberate misuse. Recently, compounds that inhibit ribosome rescue have been shown to have antibiotic activity against F. tularensis and other important pathogens. Like all bacteria that have been studied, F. tularensis uses trans-translation as the main pathway to rescue stalled ribosomes. However, unlike most bacteria, F. tularensis can survive without any of the known factors for ribosome rescue. Our work identified a F. tularensis protein, ArfT, that rescues stalled ribosomes in the absence of trans-translation using a new mechanism. These results indicate that ribosome rescue activity is essential in F. tularensis and suggest that ribosome rescue activity might be essential in all bacteria. Bacterial ribosomes frequently translate to the 3′ end of an mRNA without terminating at an in-frame stop codon. In all bacteria studied to date, these “nonstop” ribosomes are rescued using trans-translation. Genes required for trans-translation are essential in some species, but other species can survive without trans-translation because they express an alternative ribosome rescue factor, ArfA or ArfB. Francisella tularensis cells lacking trans-translation are viable, but F. tularensis does not encode ArfA or ArfB. Transposon mutagenesis followed by deep sequencing (Tn-seq) identified a new alternative ribosome rescue factor, now named ArfT. arfT can be deleted in wild-type (wt) cells but not in cells that lack trans-translation activity. Overexpression of ArfT suppresses the slow-growth phenotype in cells lacking trans-translation and counteracts growth arrest caused by trans-translation inhibitors, indicating that ArfT rescues nonstop ribosomes in vivo. Ribosome rescue assays in vitro show that ArfT promotes hydrolysis of peptidyl-tRNA on nonstop ribosomes in conjunction with F. tularensis release factors. Unlike ArfA, which requires RF2 for activity, ArfT can function with either RF1 or RF2. Overall, these results indicate that ArfT is a new alternative ribosome rescue factor with a distinct mechanism from ArfA and ArfB.
Collapse
|
99949
|
Deng SJ, Chen HY, Zeng Z, Deng S, Zhu S, Ye Z, He C, Liu ML, Huang K, Zhong JX, Xu FY, Li Q, Liu Y, Wang C, Zhao G. Nutrient Stress-Dysregulated Antisense lncRNA GLS-AS Impairs GLS-Mediated Metabolism and Represses Pancreatic Cancer Progression. Cancer Res 2018; 79:1398-1412. [PMID: 30563888 DOI: 10.1158/0008-5472.can-18-0419] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 09/25/2018] [Accepted: 12/14/2018] [Indexed: 02/05/2023]
Abstract
Cancer cells are known to undergo metabolic reprogramming, such as glycolysis and glutamine addiction, to sustain rapid proliferation and metastasis. It remains undefined whether long noncoding RNAs (lncRNA) coordinate the metabolic switch in pancreatic cancer. Here we identify a nuclear-enriched antisense lncRNA of glutaminase (GLS-AS) as a critical regulator involved in pancreatic cancer metabolism. GLS-AS was downregulated in pancreatic cancer tissues compared with noncancerous peritumor tissues. Depletion of GLS-AS promoted proliferation and invasion of pancreatic cancer cells both in vitro and in xenograft tumors of nude mice. GLS-AS inhibited GLS expression at the posttranscriptional level via formation of double stranded RNA with GLS pre-mRNA through ADAR/Dicer-dependent RNA interference. GLS-AS expression was transcriptionally downregulated by nutrient stress-induced Myc. Conversely, GLS-AS decreased Myc expression by impairing the GLS-mediated stability of Myc protein. These results imply a reciprocal feedback loop wherein Myc and GLS-AS regulate GLS overexpression during nutrient stress. Ectopic overexpression of GLS-AS inhibited proliferation and invasion of pancreatic cancer cells by repressing the Myc/GLS pathway. Moreover, expression of GLS-AS and GLS was inversely correlated in clinical samples of pancreatic cancer, while low expression of GLS-AS was associated with poor clinical outcomes. Collectively, our study implicates a novel lncRNA-mediated Myc/GLS pathway, which may serve as a metabolic target for pancreatic cancer therapy, and advances our understanding of the coupling role of lncRNA in nutrition stress and tumorigenesis.Significance: These findings show that lncRNA GLS-AS mediates a feedback loop of Myc and GLS, providing a potential therapeutic target for metabolic reprogramming in pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/7/1398/F1.large.jpg.See related commentary by Mafra and Dias, p. 1302.
Collapse
Affiliation(s)
- Shi-Jiang Deng
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng-Yu Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhu Zeng
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shichang Deng
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Zhu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng Ye
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chi He
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Liang Liu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Huang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Xin Zhong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng-Yu Xu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Li
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyou Wang
- Deparment of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Zhao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
99950
|
Kung CP, Maggi LB, Weber JD. The Role of RNA Editing in Cancer Development and Metabolic Disorders. Front Endocrinol (Lausanne) 2018; 9:762. [PMID: 30619092 PMCID: PMC6305585 DOI: 10.3389/fendo.2018.00762] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022] Open
Abstract
Numerous human diseases arise from alterations of genetic information, most notably DNA mutations. Thought to be merely the intermediate between DNA and protein, changes in RNA sequence were an afterthought until the discovery of RNA editing 30 years ago. RNA editing alters RNA sequence without altering the sequence or integrity of genomic DNA. The most common RNA editing events are A-to-I changes mediated by adenosine deaminase acting on RNA (ADAR), and C-to-U editing mediated by apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (APOBEC1). Both A-to-I and C-to-U editing were first identified in the context of embryonic development and physiological homeostasis. The role of RNA editing in human disease has only recently started to be understood. In this review, the impact of RNA editing on the development of cancer and metabolic disorders will be examined. Distinctive functions of each RNA editase that regulate either A-to-I or C-to-U editing will be highlighted in addition to pointing out important regulatory mechanisms governing these processes. The potential of developing novel therapeutic approaches through intervention of RNA editing will be explored. As the role of RNA editing in human disease is elucidated, the clinical utility of RNA editing targeted therapies will be needed. This review aims to serve as a bridge of information between past findings and future directions of RNA editing in the context of cancer and metabolic disease.
Collapse
Affiliation(s)
- Che-Pei Kung
- ICCE Institute, Washington University School of Medicine, Saint Louis, MO, United States
- Division of Molecular Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Leonard B. Maggi
- ICCE Institute, Washington University School of Medicine, Saint Louis, MO, United States
- Division of Molecular Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Jason D. Weber
- ICCE Institute, Washington University School of Medicine, Saint Louis, MO, United States
- Division of Molecular Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
- Siteman Cancer Center, Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|