1
|
Dalwani S, Metz A, Huschmann FU, Weiss MS, Wierenga RK, Venkatesan R. Crystallographic fragment-binding studies of the Mycobacterium tuberculosis trifunctional enzyme suggest binding pockets for the tails of the acyl-CoA substrates at its active sites and a potential substrate-channeling path between them. Acta Crystallogr D Struct Biol 2024; 80:605-619. [PMID: 39012716 DOI: 10.1107/s2059798324006557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
The Mycobacterium tuberculosis trifunctional enzyme (MtTFE) is an α2β2 tetrameric enzyme in which the α-chain harbors the 2E-enoyl-CoA hydratase (ECH) and 3S-hydroxyacyl-CoA dehydrogenase (HAD) active sites, and the β-chain provides the 3-ketoacyl-CoA thiolase (KAT) active site. Linear, medium-chain and long-chain 2E-enoyl-CoA molecules are the preferred substrates of MtTFE. Previous crystallographic binding and modeling studies identified binding sites for the acyl-CoA substrates at the three active sites, as well as the NAD binding pocket at the HAD active site. These studies also identified three additional CoA binding sites on the surface of MtTFE that are different from the active sites. It has been proposed that one of these additional sites could be of functional relevance for the substrate channeling (by surface crawling) of reaction intermediates between the three active sites. Here, 226 fragments were screened in a crystallographic fragment-binding study of MtTFE crystals, resulting in the structures of 16 MtTFE-fragment complexes. Analysis of the 121 fragment-binding events shows that the ECH active site is the `binding hotspot' for the tested fragments, with 41 binding events. The mode of binding of the fragments bound at the active sites provides additional insight into how the long-chain acyl moiety of the substrates can be accommodated at their proposed binding pockets. In addition, the 20 fragment-binding events between the active sites identify potential transient binding sites of reaction intermediates relevant to the possible channeling of substrates between these active sites. These results provide a basis for further studies to understand the functional relevance of the latter binding sites and to identify substrates for which channeling is crucial.
Collapse
Affiliation(s)
- Subhadra Dalwani
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Alexander Metz
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Franziska U Huschmann
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Manfred S Weiss
- Macromolecular Crystallography, Helmholtz-Zentrum Berlin, Berlin, Germany
| | - Rik K Wierenga
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rajaram Venkatesan
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
2
|
Wu S, Guo JT. Improved prediction of DNA and RNA binding proteins with deep learning models. Brief Bioinform 2024; 25:bbae285. [PMID: 38856168 PMCID: PMC11163377 DOI: 10.1093/bib/bbae285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024] Open
Abstract
Nucleic acid-binding proteins (NABPs), including DNA-binding proteins (DBPs) and RNA-binding proteins (RBPs), play important roles in essential biological processes. To facilitate functional annotation and accurate prediction of different types of NABPs, many machine learning-based computational approaches have been developed. However, the datasets used for training and testing as well as the prediction scopes in these studies have limited their applications. In this paper, we developed new strategies to overcome these limitations by generating more accurate and robust datasets and developing deep learning-based methods including both hierarchical and multi-class approaches to predict the types of NABPs for any given protein. The deep learning models employ two layers of convolutional neural network and one layer of long short-term memory. Our approaches outperform existing DBP and RBP predictors with a balanced prediction between DBPs and RBPs, and are more practically useful in identifying novel NABPs. The multi-class approach greatly improves the prediction accuracy of DBPs and RBPs, especially for the DBPs with ~12% improvement. Moreover, we explored the prediction accuracy of single-stranded DNA binding proteins and their effect on the overall prediction accuracy of NABP predictions.
Collapse
Affiliation(s)
- Siwen Wu
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, United States
| | - Jun-tao Guo
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, United States
| |
Collapse
|
3
|
Choi S, Choi SH, Bastola T, Park Y, Oh J, Kim KY, Hwang S, Miller YI, Ju WK. AIBP: A New Safeguard against Glaucomatous Neuroinflammation. Cells 2024; 13:198. [PMID: 38275823 PMCID: PMC10814024 DOI: 10.3390/cells13020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Glaucoma is a group of ocular diseases that cause irreversible blindness. It is characterized by multifactorial degeneration of the optic nerve axons and retinal ganglion cells (RGCs), resulting in the loss of vision. Major components of glaucoma pathogenesis include glia-driven neuroinflammation and impairment of mitochondrial dynamics and bioenergetics, leading to retinal neurodegeneration. In this review article, we summarize current evidence for the emerging role of apolipoprotein A-I binding protein (AIBP) as an important anti-inflammatory and neuroprotective factor in the retina. Due to its association with toll-like receptor 4 (TLR4), extracellular AIBP selectively removes excess cholesterol from the plasma membrane of inflammatory and activated cells. This results in the reduced expression of TLR4-associated, cholesterol-rich lipid rafts and the inhibition of downstream inflammatory signaling. Intracellular AIBP is localized to mitochondria and modulates mitophagy through the ubiquitination of mitofusins 1 and 2. Importantly, elevated intraocular pressure induces AIBP deficiency in mouse models and in human glaucomatous retina. AIBP deficiency leads to the activation of TLR4 in Müller glia, triggering mitochondrial dysfunction in both RGCs and Müller glia, and compromising visual function in a mouse model. Conversely, restoring AIBP expression in the retina reduces neuroinflammation, prevents RGCs death, and protects visual function. These results provide new insight into the mechanism of AIBP function in the retina and suggest a therapeutic potential for restoring retinal AIBP expression in the treatment of glaucoma.
Collapse
Affiliation(s)
- Seunghwan Choi
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tonking Bastola
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Younggun Park
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
- Department of Ophthalmology and Visual Science, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jonghyun Oh
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sinwoo Hwang
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Yury I. Miller
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| |
Collapse
|
4
|
Steensma P, Eisenhut M, Colinas M, Rosado-Souza L, Fernie AR, Weber APM, Fitzpatrick TB. PYRIDOX(AM)INE 5'-PHOSPHATE OXIDASE3 of Arabidopsis thaliana maintains carbon/nitrogen balance in distinct environmental conditions. PLANT PHYSIOLOGY 2023; 193:1433-1455. [PMID: 37453131 PMCID: PMC10517258 DOI: 10.1093/plphys/kiad411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/06/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
The identification of factors that regulate C/N utilization in plants can make a substantial contribution to optimization of plant health. Here, we explored the contribution of pyridox(am)ine 5'-phosphate oxidase3 (PDX3), which regulates vitamin B6 homeostasis, in Arabidopsis (Arabidopsis thaliana). Firstly, N fertilization regimes showed that ammonium application rescues the leaf morphological phenotype of pdx3 mutant lines but masks the metabolite perturbance resulting from impairment in utilizing soil nitrate as a source of N. Without fertilization, pdx3 lines suffered a C/N imbalance and accumulated nitrogenous compounds. Surprisingly, exploration of photorespiration as a source of endogenous N driving this metabolic imbalance, by incubation under high CO2, further exacerbated the pdx3 growth phenotype. Interestingly, the amino acid serine, critical for growth and N management, alleviated the growth phenotype of pdx3 plants under high CO2, likely due to the requirement of pyridoxal 5'-phosphate for the phosphorylated pathway of serine biosynthesis under this condition. Triggering of thermomorphogenesis by growth of plants at 28 °C (instead of 22 °C) did not appear to require PDX3 function, and we observed that the consequent drive toward C metabolism counters the C/N imbalance in pdx3. Further, pdx3 lines suffered a salicylic acid-induced defense response, probing of which unraveled that it is a protective strategy mediated by nonexpressor of pathogenesis related1 (NPR1) and improves fitness. Overall, the study demonstrates the importance of vitamin B6 homeostasis as managed by the salvage pathway enzyme PDX3 to growth in diverse environments with varying nutrient availability and insight into how plants reprogram their metabolism under such conditions.
Collapse
Affiliation(s)
- Priscille Steensma
- Department of Plant Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Marion Eisenhut
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science, Heinrich-Heine-University, Düsseldorf 40225, Germany
| | - Maite Colinas
- Department of Plant Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Laise Rosado-Souza
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Potsdam-Golm 14476, Germany
| | - Alisdair R Fernie
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Potsdam-Golm 14476, Germany
| | - Andreas P M Weber
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science, Heinrich-Heine-University, Düsseldorf 40225, Germany
| | | |
Collapse
|
5
|
Wang Z, Deng Z, Zhang W, Lou Q, Choi KS, Wei Z, Wang L, Wu J. MMSMAPlus: a multi-view multi-scale multi-attention embedding model for protein function prediction. Brief Bioinform 2023:7187109. [PMID: 37258453 DOI: 10.1093/bib/bbad201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/16/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023] Open
Abstract
Protein is the most important component in organisms and plays an indispensable role in life activities. In recent years, a large number of intelligent methods have been proposed to predict protein function. These methods obtain different types of protein information, including sequence, structure and interaction network. Among them, protein sequences have gained significant attention where methods are investigated to extract the information from different views of features. However, how to fully exploit the views for effective protein sequence analysis remains a challenge. In this regard, we propose a multi-view, multi-scale and multi-attention deep neural model (MMSMA) for protein function prediction. First, MMSMA extracts multi-view features from protein sequences, including one-hot encoding features, evolutionary information features, deep semantic features and overlapping property features based on physiochemistry. Second, a specific multi-scale multi-attention deep network model (MSMA) is built for each view to realize the deep feature learning and preliminary classification. In MSMA, both multi-scale local patterns and long-range dependence from protein sequences can be captured. Third, a multi-view adaptive decision mechanism is developed to make a comprehensive decision based on the classification results of all the views. To further improve the prediction performance, an extended version of MMSMA, MMSMAPlus, is proposed to integrate homology-based protein prediction under the framework of multi-view deep neural model. Experimental results show that the MMSMAPlus has promising performance and is significantly superior to the state-of-the-art methods. The source code can be found at https://github.com/wzy-2020/MMSMAPlus.
Collapse
Affiliation(s)
- Zhongyu Wang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Zhaohong Deng
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Wei Zhang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Qiongdan Lou
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | | | - Zhisheng Wei
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| | - Lei Wang
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| | - Jing Wu
- National Key Laboratory of Food Science and Resource Mining, Jiangnan University, Wuxi, China
| |
Collapse
|
6
|
Zegarra V, Mais CN, Freitag J, Bange G. The mysterious diadenosine tetraphosphate (AP4A). MICROLIFE 2023; 4:uqad016. [PMID: 37223742 PMCID: PMC10148737 DOI: 10.1093/femsml/uqad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/15/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2023]
Abstract
Dinucleoside polyphosphates, a class of nucleotides found amongst all the Trees of Life, have been gathering a lot of attention in the past decades due to their putative role as cellular alarmones. In particular, diadenosine tetraphosphate (AP4A) has been widely studied in bacteria facing various environmental challenges and has been proposed to be important for ensuring cellular survivability through harsh conditions. Here, we discuss the current understanding of AP4A synthesis and degradation, protein targets, their molecular structure where possible, and insights into the molecular mechanisms of AP4A action and its physiological consequences. Lastly, we will briefly touch on what is known with regards to AP4A beyond the bacterial kingdom, given its increasing appearance in the eukaryotic world. Altogether, the notion that AP4A is a conserved second messenger in organisms ranging from bacteria to humans and is able to signal and modulate cellular stress regulation seems promising.
Collapse
Affiliation(s)
- Victor Zegarra
- Department of Chemistry and Center for Synthetic Microbiology, Philipps University Marburg, Marburg 35043, Germany
| | - Christopher-Nils Mais
- Department of Chemistry and Center for Synthetic Microbiology, Philipps University Marburg, Marburg 35043, Germany
| | - Johannes Freitag
- Department of Biology, Philipps University Marburg, Marburg 35043, Germany
| | - Gert Bange
- Corresponding author. Karl-von-Frisch Strasse 14, 35043 Marburg, Germany. E-mail:
| |
Collapse
|
7
|
Angira D, Chaudhary S, Abiramasundari A, Thiruvenkatam V. To Explore the Binding Affinity of Human γ-Secretase Activating Protein (GSAP) Isoform 4 with APP-C99 Peptides. ACS OMEGA 2023; 8:13435-13443. [PMID: 37065030 PMCID: PMC10099435 DOI: 10.1021/acsomega.3c01117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
γ-Secretase activating protein (GSAP) is known to play an important role in the β-amyloid pathway. It acts as a modulator and accentuates the truncation of the amyloid precursor protein C-99 fragment through the γ-secretase complex. GSAP has four isoforms, out of which canonical isoform 1, a 16 kDa C-terminal portion, has been extensively studied, whereas the function of other three isoforms remains unknown. Here, we explore the GSAP isoform 4 (GSAP_I4) expression and purification from inclusion bodies followed by the refolding of the protein. The secondary structure of GSAP_I4 is predicted using circular dichroism. The protein is further characterized by western blotting and mass spectroscopy analysis. Additionally, biochemical assays and in silico molecular docking and molecular simulation are performed to investigate the binding of GSAP_I4 and APP-C99 peptide fragments. The results reflect that although GSAP_I1 and GSAP_I4 share high sequence similarity, the isoform 4 does not show any affinity toward APP-C99 peptide fragments. This hints toward the fact that GSAP_I4 might have a different role in the living system that is yet unexplored.
Collapse
Affiliation(s)
- Deekshi Angira
- Discipline
of Chemistry, Indian Institute of Technology
Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Sonali Chaudhary
- Discipline
of Chemistry, Indian Institute of Technology
Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Arumugam Abiramasundari
- Discipline
of Biological Engineering, Indian Institute
of Technology Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Vijay Thiruvenkatam
- Discipline
of Biological Engineering, Indian Institute
of Technology Gandhinagar, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
8
|
Kim JD, Zhou T, Zhang A, Li S, Gupte AA, Hamilton DJ, Fang L. AIBP Regulates Metabolism of Ketone and Lipids but Not Mitochondrial Respiration. Cells 2022; 11:cells11223643. [PMID: 36429071 PMCID: PMC9688289 DOI: 10.3390/cells11223643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence indicates that the APOA1 binding protein (AIBP)-a secreted protein-plays a profound role in lipid metabolism. Interestingly, AIBP also functions as an NAD(P)H-hydrate epimerase to catalyze the interconversion of NAD(P)H hydrate [NAD(P)HX] epimers and is renamed as NAXE. Thus, we call it NAXE hereafter. We investigated its role in NAD(P)H-involved metabolism in murine cardiomyocytes, focusing on the metabolism of hexose, lipids, and amino acids as well as mitochondrial redox function. Unbiased metabolite profiling of cardiac tissue shows that NAXE knockout markedly upregulates the ketone body 3-hydroxybutyric acid (3-HB) and increases or trends increasing lipid-associated metabolites cholesterol, α-linolenic acid and deoxycholic acid. Paralleling greater ketone levels, ChemRICH analysis of the NAXE-regulated metabolites shows reduced abundance of hexose despite similar glucose levels in control and NAXE-deficient blood. NAXE knockout reduces cardiac lactic acid but has no effect on the content of other NAD(P)H-regulated metabolites, including those associated with glucose metabolism, the pentose phosphate pathway, or Krebs cycle flux. Although NAXE is present in mitochondria, it has no apparent effect on mitochondrial oxidative phosphorylation. Instead, we detected more metabolites that can potentially improve cardiac function (3-HB, adenosine, and α-linolenic acid) in the Naxe-/- heart; these mice also perform better in aerobic exercise. Our data reveal a new role of NAXE in cardiac ketone and lipid metabolism.
Collapse
Affiliation(s)
- Jun-dae Kim
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
| | - Teng Zhou
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, 6550 Fannin St., Houston, TX 77030, USA
| | - Shumin Li
- Center for Bioenergetics, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
| | - Anisha A. Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, 6550 Fannin St., Houston, TX 77030, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, 6550 Fannin St., Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, 407 E 61st St., New York, NY 10065, USA
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin St., Houston, TX 77030, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, 6550 Fannin St., Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, 407 E 61st St., New York, NY 10065, USA
- Correspondence: ; Tel.: +713-363-9012; Fax: +713-363-9782
| |
Collapse
|
9
|
Guo JT, Malik F. Single-Stranded DNA Binding Proteins and Their Identification Using Machine Learning-Based Approaches. Biomolecules 2022; 12:biom12091187. [PMID: 36139026 PMCID: PMC9496475 DOI: 10.3390/biom12091187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/11/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Single-stranded DNA (ssDNA) binding proteins (SSBs) are critical in maintaining genome stability by protecting the transient existence of ssDNA from damage during essential biological processes, such as DNA replication and gene transcription. The single-stranded region of telomeres also requires protection by ssDNA binding proteins from being attacked in case it is wrongly recognized as an anomaly. In addition to their critical roles in genome stability and integrity, it has been demonstrated that ssDNA and SSB-ssDNA interactions play critical roles in transcriptional regulation in all three domains of life and viruses. In this review, we present our current knowledge of the structure and function of SSBs and the structural features for SSB binding specificity. We then discuss the machine learning-based approaches that have been developed for the prediction of SSBs from double-stranded DNA (dsDNA) binding proteins (DSBs).
Collapse
|
10
|
Duan M, Chen H, Yin L, Zhu X, Novák P, Lv Y, Zhao G, Yin K. Mitochondrial apolipoprotein A-I binding protein alleviates atherosclerosis by regulating mitophagy and macrophage polarization. Cell Commun Signal 2022; 20:60. [PMID: 35525979 PMCID: PMC9077873 DOI: 10.1186/s12964-022-00858-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/05/2022] [Indexed: 12/22/2022] Open
Abstract
Apolipoprotein A-I binding protein (AIBP), a secreted protein, has been shown to play a pivotal role in the development of atherosclerosis. The function of intracellular AIBP, however, is not yet well characterized. Here, we found that AIBP is abundantly expressed within human and mouse atherosclerotic lesions and exhibits a distinct localization in the inner membrane of mitochondria in macrophages. Bone marrow-specific AIBP deficiency promotes the progression of atherosclerosis and increases macrophage infiltration and inflammation in low-density lipoprotein receptor-deficient (LDLR-/-) mice. Specifically, the lack of mitochondrial AIBP leads to mitochondrial metabolic disorders, thereby reducing the formation of mitophagy by promoting the cleavage of PTEN-induced putative kinase 1 (PINK1). With the reduction in mitochondrial autophagy, macrophages polarize to the M1 proinflammatory phenotype, which further promotes the development of atherosclerosis. Based on these results, mitochondrial AIBP in macrophages performs an antiatherosclerotic role by regulating of PINK1-dependent mitophagy and M1/M2 polarization. Video Abstract.
Collapse
Affiliation(s)
- Meng Duan
- Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, 541100 Guangxi China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi China
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Hainan Chen
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Linjie Yin
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi China
| | - Yuncheng Lv
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, 511518 Guangdong China
| | - Kai Yin
- Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin, 541100 Guangxi China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi China
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
11
|
Kim JD, Zhu L, Sun Q, Fang L. Systemic metabolite profiling reveals sexual dimorphism of AIBP control of metabolism in mice. PLoS One 2021; 16:e0248964. [PMID: 33793635 PMCID: PMC8016339 DOI: 10.1371/journal.pone.0248964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/08/2021] [Indexed: 01/04/2023] Open
Abstract
Emerging studies indicate that APOA-I binding protein (AIBP) is a secreted protein and functions extracellularly to promote cellular cholesterol efflux, thereby disrupting lipid rafts on the plasma membrane. AIBP is also present in the mitochondria and acts as an epimerase, facilitating the repair of dysfunctional hydrated NAD(P)H, known as NAD(P)H(X). Importantly, AIBP deficiency contributes to lethal neurometabolic disorder, reminiscent of the Leigh syndrome in humans. Whereas cyclic NADPHX production is proposed to be the underlying cause, we hypothesize that an unbiased metabolic profiling may: 1) reveal new clues for the lethality, e.g., changes of mitochondrial metabolites., and 2) identify metabolites associated with new AIBP functions. To this end, we performed unbiased and profound metabolic studies of plasma obtained from adult AIBP knockout mice and control littermates of both genders. Our systemic metabolite profiling, encompassing 9 super pathways, identified a total of 640 compounds. Our studies demonstrate a surprising sexual dimorphism of metabolites affected by AIBP deletion, with more statistically significant changes in the AIBP knockout female vs male when compared with the corresponding controls. AIBP knockout trends to reduce cholesterol but increase the bile acid precursor 7-HOCA in female but not male. Complex lipids, phospholipids, sphingomyelin and plasmalogens were reduced, while monoacylglycerol, fatty acids and the lipid soluble vitamins E and carotene diol were elevated in AIBP knockout female but not male. NAD metabolites were not significantly different in AIBP knockout vs control mice but differed for male vs female mice. Metabolites associated with glycolysis and the Krebs cycle were unchanged by AIBP knockout. Importantly, polyamine spermidine, critical for many cellular functions including cerebral cortex synapses, was reduced in male but not female AIBP knockout. This is the first report of a systemic metabolite profile of plasma samples from AIBP knockout mice, and provides a metabolic basis for future studies of AIBP regulation of cellular metabolism and the pathophysiological presentation of AIBP deficiency in patients.
Collapse
Affiliation(s)
- Jun-dae Kim
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States of America
| | - Lingping Zhu
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States of America
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Quan Sun
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States of America
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States of America
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States of America
- Weill Cornell Medical College, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
12
|
Choi SH, Agatisa-Boyle C, Gonen A, Kim A, Kim J, Alekseeva E, Tsimikas S, Miller YI. Intracellular AIBP (Apolipoprotein A-I Binding Protein) Regulates Oxidized LDL (Low-Density Lipoprotein)-Induced Mitophagy in Macrophages. Arterioscler Thromb Vasc Biol 2021; 41:e82-e96. [PMID: 33356389 PMCID: PMC8105271 DOI: 10.1161/atvbaha.120.315485] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Atherosclerotic lesions are often characterized by accumulation of OxLDL (oxidized low-density lipoprotein), which is associated with vascular inflammation and lesion vulnerability to rupture. Extracellular AIBP (apolipoprotein A-I binding protein; encoded by APOA1BP gene), when secreted, promotes cholesterol efflux and regulates lipid rafts dynamics, but its role as an intracellular protein in mammalian cells remains unknown. The aim of this work was to determine the function of intracellular AIBP in macrophages exposed to OxLDL and in atherosclerotic lesions. Approach and Results: Using a novel monoclonal antibody against human and mouse AIBP, which are highly homologous, we demonstrated robust AIBP expression in human and mouse atherosclerotic lesions. We observed significantly reduced autophagy in bone marrow-derived macrophages, isolated from Apoa1bp-/- compared with wild-type mice, which were exposed to OxLDL. In atherosclerotic lesions from Apoa1bp-/- mice subjected to Ldlr knockdown and fed a Western diet, autophagy was reduced, whereas apoptosis was increased, when compared with that in wild-type mice. AIBP expression was necessary for efficient control of reactive oxygen species and cell death and for mitochondria quality control in macrophages exposed to OxLDL. Mitochondria-localized AIBP, via its N-terminal domain, associated with E3 ubiquitin-protein ligase PARK2 (Parkin), MFN (mitofusin)1, and MFN2, but not BNIP3 (Bcl2/adenovirus E1B 19-kDa-interacting protein-3), and regulated ubiquitination of MFN1 and MFN2, key components of mitophagy. CONCLUSIONS These data suggest that intracellular AIBP is a new regulator of autophagy in macrophages. Mitochondria-localized AIBP augments mitophagy and participates in mitochondria quality control, protecting macrophages against cell death in the context of atherosclerosis.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Colin Agatisa-Boyle
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Ayelet Gonen
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Alisa Kim
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Jungsu Kim
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Elena Alekseeva
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Sotirios Tsimikas
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| | - Yury I. Miller
- Department of Medicine University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
13
|
Qiu X, Luo J, Fang L. AIBP, Angiogenesis, Hematopoiesis, and Atherogenesis. Curr Atheroscler Rep 2020; 23:1. [PMID: 33230630 DOI: 10.1007/s11883-020-00899-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW The goal of this manuscript is to summarize the current understanding of the secreted APOA1 binding protein (AIBP), encoded by NAXE, in angiogenesis, hematopoiesis, and inflammation. The studies on AIBP illustrate a critical connection between lipid metabolism and the aforementioned endothelial and immune cell biology. RECENT FINDINGS AIBP dictates both developmental processes such as angiogenesis and hematopoiesis, and pathological events such as inflammation, tumorigenesis, and atherosclerosis. Although cholesterol efflux dictates AIBP-mediated lipid raft disruption in many of the cell types, recent studies document cholesterol efflux-independent mechanism involving Cdc42-mediated cytoskeleton remodeling in macrophages. AIBP disrupts lipid rafts and impairs raft-associated VEGFR2 but facilitates non-raft-associated NOTCH1 signaling. Furthermore, AIBP can induce cholesterol biosynthesis gene SREBP2 activation, which in turn transactivates NOTCH1 and supports specification of hematopoietic stem and progenitor cells (HSPCs). In addition, AIBP also binds TLR4 and represses TLR4-mediated inflammation. In this review, we summarize the latest research on AIBP, focusing on its role in cholesterol metabolism and the attendant effects on lipid raft-regulated VEGFR2 and non-raft-associated NOTCH1 activation in angiogenesis, SREBP2-upregulated NOTCH1 signaling in hematopoiesis, and TLR4 signaling in inflammation and atherogenesis. We will discuss its potential therapeutic applications in angiogenesis and inflammation due to selective targeting of activated cells.
Collapse
Affiliation(s)
- Xueting Qiu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA
| | - Jingmin Luo
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Department of Obstetrics and Gynecology, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
14
|
Qing J, Zhang Z, Novák P, Zhao G, Yin K. Mitochondrial metabolism in regulating macrophage polarization: an emerging regulator of metabolic inflammatory diseases. Acta Biochim Biophys Sin (Shanghai) 2020; 52:917-926. [PMID: 32785581 DOI: 10.1093/abbs/gmaa081] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Indexed: 12/24/2022] Open
Abstract
As a major type of immune cells with heterogeneity and plasticity, macrophages are classically divided into inflammatory (M1) and alternative/anti-inflammatory (M2) types and play a crucial role in the progress of the inflammatory diseases. Recent studies have shown that metabolism is an important determinant of macrophage phenotype. Mitochondria, one of the most important compartments involving cell metabolism, are closely associated with the regulation of cell functions. In most types of cell, mitochondrial oxidative phosphorylation (OXPHOS) is the primary mode of cellular energy production. However, mitochondrial OXPHOS is inhibited in activated M1 macrophages, rendering them unable to be converted into M2 phenotype. Thus, mitochondrial metabolism is a crucial regulator in macrophage functions. This review summarizes the roles of mitochondria in macrophage polarization and analyzes the molecular mechanisms underlying mitochondrial metabolism and function, which may provide new approaches for the treatment of metabolic inflammatory diseases.
Collapse
Affiliation(s)
- Jina Qing
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zizhen Zhang
- School of Medicine, Hunan Polytechnic of Environment and Biology, Hengyang 421001, China
| | - Petr Novák
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan 511518, China
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
15
|
Choi SH, Kim KY, Perkins GA, Phan S, Edwards G, Xia Y, Kim J, Skowronska-Krawczyk D, Weinreb RN, Ellisman MH, Miller YI, Ju WK. AIBP protects retinal ganglion cells against neuroinflammation and mitochondrial dysfunction in glaucomatous neurodegeneration. Redox Biol 2020; 37:101703. [PMID: 32896719 PMCID: PMC7484594 DOI: 10.1016/j.redox.2020.101703] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/12/2020] [Accepted: 08/22/2020] [Indexed: 01/10/2023] Open
Abstract
Glaucoma is a leading cause of blindness worldwide in individuals 60 years of age and older. Despite its high prevalence, the factors contributing to glaucoma progression are currently not well characterized. Glia-driven neuroinflammation and mitochondrial dysfunction play critical roles in glaucomatous neurodegeneration. Here, we demonstrated that elevated intraocular pressure (IOP) significantly decreased apolipoprotein A-I binding protein (AIBP; gene name Apoa1bp) in retinal ganglion cells (RGCs), but resulted in upregulation of TLR4 and IL-1β expression in Müller glia endfeet. Apoa1bp-/- mice had impaired visual function and Müller glia characterized by upregulated TLR4 activity, impaired mitochondrial network and function, increased oxidative stress and induced inflammatory responses. We also found that AIBP deficiency compromised mitochondrial network and function in RGCs and exacerbated RGC vulnerability to elevated IOP. Administration of recombinant AIBP prevented RGC death and inhibited inflammatory responses and cytokine production in Müller glia in vivo. These findings indicate that AIBP protects RGCs against glia-driven neuroinflammation and mitochondrial dysfunction in glaucomatous neurodegeneration and suggest that recombinant AIBP may be a potential therapeutic agent for glaucoma.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sébastien Phan
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Genea Edwards
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yining Xia
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jungsu Kim
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology, Biophysics & Ophthalmology, University of California Irvine, Irvine, CA, 92697, USA
| | - Robert N Weinreb
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yury I Miller
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Low H, Mukhamedova N, Capettini LDSA, Xia Y, Carmichael I, Cody SH, Huynh K, Ditiatkovski M, Ohkawa R, Bukrinsky M, Meikle PJ, Choi SH, Field S, Miller YI, Sviridov D. Cholesterol Efflux-Independent Modification of Lipid Rafts by AIBP (Apolipoprotein A-I Binding Protein). Arterioscler Thromb Vasc Biol 2020; 40:2346-2359. [PMID: 32787522 PMCID: PMC7530101 DOI: 10.1161/atvbaha.120.315037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE AIBP (apolipoprotein A-I binding protein) is an effective and selective regulator of lipid rafts modulating many metabolic pathways originating from the rafts, including inflammation. The mechanism of action was suggested to involve stimulation by AIBP of cholesterol efflux, depleting rafts of cholesterol, which is essential for lipid raft integrity. Here we describe a different mechanism contributing to the regulation of lipid rafts by AIBP. Approach and Results: We demonstrate that modulation of rafts by AIBP may not exclusively depend on the rate of cholesterol efflux or presence of the key regulator of the efflux, ABCA1 (ATP-binding cassette transporter A-I). AIBP interacted with phosphatidylinositol 3-phosphate, which was associated with increased abundance and activation of Cdc42 and rearrangement of the actin cytoskeleton. Cytoskeleton rearrangement was accompanied with reduction of the abundance of lipid rafts, without significant changes in the lipid composition of the rafts. The interaction of AIBP with phosphatidylinositol 3-phosphate was blocked by AIBP substrate, NADPH (nicotinamide adenine dinucleotide phosphate), and both NADPH and silencing of Cdc42 interfered with the ability of AIBP to regulate lipid rafts and cholesterol efflux. CONCLUSIONS Our findings indicate that an underlying mechanism of regulation of lipid rafts by AIBP involves PIP-dependent rearrangement of the cytoskeleton.
Collapse
Affiliation(s)
- Hann Low
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.)
| | - Nigora Mukhamedova
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.)
| | - Luciano Dos Santos Aggum Capettini
- Department of Medicine, University of California San Diego, La Jolla (L.d.S.A.C., Y.X., S.-H.C., S.F., Y.I.M.).,Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil (L.d.S.A.C.)
| | - Yining Xia
- Department of Medicine, University of California San Diego, La Jolla (L.d.S.A.C., Y.X., S.-H.C., S.F., Y.I.M.)
| | - Irena Carmichael
- Department of Monash Micro Imaging, Monash University, Melbourne, VIC, Australia (I.C., S.H.C.)
| | - Stephen H Cody
- Department of Monash Micro Imaging, Monash University, Melbourne, VIC, Australia (I.C., S.H.C.)
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.)
| | - Michael Ditiatkovski
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.)
| | - Ryunosuke Ohkawa
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.).,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan (R.O.)
| | - Michael Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, DC (M.B.)
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.)
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla (L.d.S.A.C., Y.X., S.-H.C., S.F., Y.I.M.)
| | - Seth Field
- Department of Medicine, University of California San Diego, La Jolla (L.d.S.A.C., Y.X., S.-H.C., S.F., Y.I.M.)
| | - Yury I Miller
- Department of Medicine, University of California San Diego, La Jolla (L.d.S.A.C., Y.X., S.-H.C., S.F., Y.I.M.)
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (H.L., N.M., K.H., M.D., R.O., P.J.M., D.S.).,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia (D.S.)
| |
Collapse
|
17
|
NPF:network propagation for protein function prediction. BMC Bioinformatics 2020; 21:355. [PMID: 32787776 PMCID: PMC7430911 DOI: 10.1186/s12859-020-03663-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/14/2020] [Indexed: 11/29/2022] Open
Abstract
Background The accurate annotation of protein functions is of great significance in elucidating the phenomena of life, treating disease and developing new medicines. Various methods have been developed to facilitate the prediction of these functions by combining protein interaction networks (PINs) with multi-omics data. However, it is still challenging to make full use of multiple biological to improve the performance of functions annotation. Results We presented NPF (Network Propagation for Functions prediction), an integrative protein function predicting framework assisted by network propagation and functional module detection, for discovering interacting partners with similar functions to target proteins. NPF leverages knowledge of the protein interaction network architecture and multi-omics data, such as domain annotation and protein complex information, to augment protein-protein functional similarity in a propagation manner. We have verified the great potential of NPF for accurately inferring protein functions. According to the comprehensive evaluation of NPF, it delivered a better performance than other competing methods in terms of leave-one-out cross-validation and ten-fold cross validation. Conclusions We demonstrated that network propagation, together with multi-omics data, can both discover more partners with similar function, and is unconstricted by the “small-world” feature of protein interaction networks. We conclude that the performance of function prediction depends greatly on whether we can extract and exploit proper functional information of similarity from protein correlations.
Collapse
|
18
|
Shabalin IG, Porebski PJ, Minor W. Refining the macromolecular model - achieving the best agreement with the data from X-ray diffraction experiment. CRYSTALLOGR REV 2018; 24:236-262. [PMID: 30416256 DOI: 10.1080/0889311x.2018.1521805] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Refinement of macromolecular X-ray crystal structures involves using complex software with hundreds of different settings. The complexity of underlying concepts and the sheer amount sof instructions may make it difficult for less experienced crystallographers to achieve optimal results in their refinements. This tutorial review offers guidelines for choosing the best settings for the reciprocal-space refinement of macromolecular models and provides practical tips for manual model correction. To help aspiring crystallographers navigate the process, some of the most practically important concepts of protein structure refinement are described. Among the topics covered are the use and purpose of R-free, geometrical restraints, restraints on atomic displacement parameters (ADPs), refinement weights, various parametrizations of ADPs (full anisotropic refinement and TLS), and omit maps. We also give practical tips for manual model correction in Coot, modelling of side-chains with poor or missing density, and ligand identification, fitting, and refinement.
Collapse
Affiliation(s)
- Ivan G Shabalin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), Charlottesville, VA, 22908, United States
| | - Przemyslaw J Porebski
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), Charlottesville, VA, 22908, United States
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), Charlottesville, VA, 22908, United States
| |
Collapse
|
19
|
Choi SH, Wallace AM, Schneider DA, Burg E, Kim J, Alekseeva E, Ubags ND, Cool CD, Fang L, Suratt BT, Miller YI. AIBP augments cholesterol efflux from alveolar macrophages to surfactant and reduces acute lung inflammation. JCI Insight 2018; 3:120519. [PMID: 30135304 DOI: 10.1172/jci.insight.120519] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by an excessive pulmonary inflammatory response. Removal of excess cholesterol from the plasma membrane of inflammatory cells helps reduce their activation. The secreted apolipoprotein A-I binding protein (AIBP) has been shown to augment cholesterol efflux from endothelial cells to the plasma lipoprotein HDL. Here, we find that AIBP was expressed in inflammatory cells in the human lung and was secreted into the bronchoalveolar space in mice subjected to inhalation of LPS. AIBP bound surfactant protein B and increased cholesterol efflux from alveolar macrophages to calfactant, a therapeutic surfactant formulation. In vitro, AIBP in the presence of surfactant reduced LPS-induced p65, ERK1/2 and p38 phosphorylation, and IL-6 secretion by alveolar macrophages. In vivo, inhalation of AIBP significantly reduced LPS-induced airspace neutrophilia, alveolar capillary leak, and secretion of IL-6. These results suggest that, similar to HDL in plasma, surfactant serves as a cholesterol acceptor in the lung. Furthermore, lung injury increases pulmonary AIBP expression, which likely serves to promote cholesterol efflux to surfactant and reduce inflammation.
Collapse
Affiliation(s)
- Soo-Ho Choi
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Aaron M Wallace
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | | | - Elianne Burg
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Jungsu Kim
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | - Niki Dj Ubags
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Carlyne D Cool
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, and.,Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist, Houston, Texas, USA
| | - Benjamin T Suratt
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Yury I Miller
- Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
20
|
Komárek J, Ivanov Kavková E, Houser J, Horáčková A, Ždánská J, Demo G, Wimmerová M. Structure and properties of AB21, a novelAgaricus bisporusprotein with structural relation to bacterial pore-forming toxins. Proteins 2018; 86:897-911. [DOI: 10.1002/prot.25522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Jan Komárek
- Central European Institute of Technology, Masaryk University, Kamenice 5; Brno 62500 Czech Republic
- National Centre for Biomolecular Research; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| | - Eva Ivanov Kavková
- Department of Biochemistry; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| | - Josef Houser
- Central European Institute of Technology, Masaryk University, Kamenice 5; Brno 62500 Czech Republic
- National Centre for Biomolecular Research; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| | - Aneta Horáčková
- Department of Biochemistry; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| | - Jitka Ždánská
- Central European Institute of Technology, Masaryk University, Kamenice 5; Brno 62500 Czech Republic
| | - Gabriel Demo
- Central European Institute of Technology, Masaryk University, Kamenice 5; Brno 62500 Czech Republic
- National Centre for Biomolecular Research; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| | - Michaela Wimmerová
- Central European Institute of Technology, Masaryk University, Kamenice 5; Brno 62500 Czech Republic
- National Centre for Biomolecular Research; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
- Department of Biochemistry; Faculty of Science, Masaryk University, Kotlarska 2; Brno 61137 Czech Republic
| |
Collapse
|
21
|
Evidence that the metabolite repair enzyme NAD(P)HX epimerase has a moonlighting function. Biosci Rep 2018; 38:BSR20180223. [PMID: 29654173 PMCID: PMC5938422 DOI: 10.1042/bsr20180223] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 11/25/2022] Open
Abstract
NAD(P)H-hydrate epimerase (EC 5.1.99.6) is known to help repair NAD(P)H hydrates (NAD(P)HX), which are damage products existing as R and S epimers. The S epimer is reconverted to NAD(P)H by a dehydratase; the epimerase facilitates epimer interconversion. Epimerase deficiency in humans causes a lethal disorder attributed to NADHX accumulation. However, bioinformatic evidence suggest caution about this attribution by predicting that the epimerase has a second function connected to vitamin B6 (pyridoxal 5′-phosphate and related compounds). Specifically, (i) the epimerase is fused to a B6 salvage enzyme in plants, (ii) epimerase genes cluster on the chromosome with B6-related genes in bacteria, and (iii) epimerase and B6-related genes are coexpressed in yeast and Arabidopsis. The predicted second function was explored in Escherichia coli, whose epimerase and dehydratase are fused and encoded by yjeF. The putative NAD(P)HX epimerase active site has a conserved lysine residue (K192 in E. coli YjeF). Changing this residue to alanine cut in vitro epimerase activity by ≥95% but did not affect dehydratase activity. Mutant cells carrying the K192A mutation had essentially normal NAD(P)HX dehydratase activity and NAD(P)HX levels, showing that the mutation had little impact on NAD(P)HX repair in vivo. However, these cells showed metabolome changes, particularly in amino acids, which exceeded those in cells lacking the entire yjeF gene. The K192A mutant cells also had reduced levels of ‘free’ (i.e. weakly bound or unbound) pyridoxal 5'-phosphate. These results provide circumstantial evidence that the epimerase has a metabolic function beyond NAD(P)HX repair and that this function involves vitamin B6.
Collapse
|
22
|
Schneider DA, Choi SH, Agatisa-Boyle C, Zhu L, Kim J, Pattison J, Sears DD, Gordts PLSM, Fang L, Miller YI. AIBP protects against metabolic abnormalities and atherosclerosis. J Lipid Res 2018; 59:854-863. [PMID: 29559522 PMCID: PMC5928435 DOI: 10.1194/jlr.m083618] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Apolipoprotein A-I binding protein (AIBP) has been shown to augment cholesterol efflux from endothelial cells and macrophages. In zebrafish and mice, AIBP-mediated regulation of cholesterol levels in the plasma membrane of endothelial cells controls angiogenesis. The goal of this work was to evaluate metabolic changes and atherosclerosis in AIBP loss-of-function and gain-of-function animal studies. Here, we show that Apoa1bp-/-Ldlr-/- mice fed a high-cholesterol, high-fat diet had exacerbated weight gain, liver steatosis, glucose intolerance, hypercholesterolemia, hypertriglyceridemia, and larger atherosclerotic lesions compared with Ldlr-/- mice. Feeding Apoa1bp-/-Ldlr-/- mice a high-cholesterol, normal-fat diet did not result in significant differences in lipid levels or size of atherosclerotic lesions from Ldlr-/- mice. Conversely, adeno-associated virus-mediated overexpression of AIBP reduced hyperlipidemia and atherosclerosis in high-cholesterol, high-fat diet-fed Ldlr-/- mice. Injections of recombinant AIBP reduced aortic inflammation in Ldlr-/- mice fed a short high-cholesterol, high-fat diet. Conditional overexpression of AIBP in zebrafish also reduced diet-induced vascular lipid accumulation. In experiments with isolated macrophages, AIBP facilitated cholesterol efflux to HDL, reduced lipid rafts content, and inhibited inflammatory responses to lipopolysaccharide.jlr Our data demonstrate that AIBP confers protection against diet-induced metabolic abnormalities and atherosclerosis.
Collapse
Affiliation(s)
- Dina A Schneider
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Soo-Ho Choi
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Colin Agatisa-Boyle
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Laurence Zhu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030
| | - Jungsu Kim
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Jennifer Pattison
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Dorothy D Sears
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093.,Family Medicine and Public Health, University of California at San Diego, La Jolla, CA 92093
| | - Philip L S M Gordts
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030
| | - Yury I Miller
- Departments of Medicine University of California at San Diego, La Jolla, CA 92093
| |
Collapse
|
23
|
Abstract
Recent advances in high-throughput structure determination and computational protein structure prediction have significantly enriched the universe of protein structure. However, there is still a large gap between the number of available protein structures and that of proteins with annotated function in high accuracy. Computational structure-based protein function prediction has emerged to reduce this knowledge gap. The identification of a ligand binding site and its structure is critical to the determination of a protein's molecular function. We present a computational methodology for predicting small molecule ligand binding site and ligand structure using G-LoSA, our protein local structure alignment and similarity measurement tool. All the computational procedures described here can be easily implemented using G-LoSA Toolkit, a package of standalone software programs and preprocessed PDB structure libraries. G-LoSA and G-LoSA Toolkit are freely available to academic users at http://compbio.lehigh.edu/GLoSA . We also illustrate a case study to show the potential of our template-based approach harnessing G-LoSA for protein function prediction.
Collapse
Affiliation(s)
- Hui Sun Lee
- Department of Biological Sciences and Bioengineering Program, Lehigh University, Bethlehem, PA, 18015, USA.
| | - Wonpil Im
- Department of Biological Sciences and Bioengineering Program, Lehigh University, Bethlehem, PA, 18015, USA.
| |
Collapse
|
24
|
Ellens KW, Christian N, Singh C, Satagopam VP, May P, Linster CL. Confronting the catalytic dark matter encoded by sequenced genomes. Nucleic Acids Res 2017; 45:11495-11514. [PMID: 29059321 PMCID: PMC5714238 DOI: 10.1093/nar/gkx937] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 10/03/2017] [Indexed: 01/02/2023] Open
Abstract
The post-genomic era has provided researchers with a deluge of protein sequences. However, a significant fraction of the proteins encoded by sequenced genomes remains without an identified function. Here, we aim at determining how many enzymes of uncertain or unknown function are still present in the Saccharomyces cerevisiae and human proteomes. Using information available in the Swiss-Prot, BRENDA and KEGG databases in combination with a Hidden Markov Model-based method, we estimate that >600 yeast and 2000 human proteins (>30% of their proteins of unknown function) are enzymes whose precise function(s) remain(s) to be determined. This illustrates the impressive scale of the ‘unknown enzyme problem’. We extensively review classical biochemical as well as more recent systematic experimental and computational approaches that can be used to support enzyme function discovery research. Finally, we discuss the possible roles of the elusive catalysts in light of recent developments in the fields of enzymology and metabolism as well as the significance of the unknown enzyme problem in the context of metabolic modeling, metabolic engineering and rare disease research.
Collapse
Affiliation(s)
- Kenneth W Ellens
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Nils Christian
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Charandeep Singh
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Venkata P Satagopam
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
25
|
Earl LA, Falconieri V, Milne JL, Subramaniam S. Cryo-EM: beyond the microscope. Curr Opin Struct Biol 2017; 46:71-78. [PMID: 28646653 PMCID: PMC5683925 DOI: 10.1016/j.sbi.2017.06.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/31/2017] [Accepted: 06/06/2017] [Indexed: 01/18/2023]
Abstract
The pace at which cryo-EM is being adopted as a mainstream tool in structural biology has continued unabated over the past year. Initial successes in obtaining near-atomic resolution structures with cryo-EM were enabled to a large extent by advances in microscope and detector technology. Here, we review some of the complementary technical improvements that are helping sustain the cryo-EM revolution. We highlight advances in image processing that permit high resolution structure determination even in the presence of structural and conformational heterogeneity. We also review selected examples where biochemical strategies for membrane protein stabilization facilitate cryo-EM structure determination, and discuss emerging approaches for further improving the preparation of reliable plunge-frozen specimens.
Collapse
Affiliation(s)
- Lesley A Earl
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Veronica Falconieri
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline Ls Milne
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sriram Subramaniam
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Zhang M, Li L, Xie W, Wu JF, Yao F, Tan YL, Xia XD, Liu XY, Liu D, Lan G, Zeng MY, Gong D, Cheng HP, Huang C, Zhao ZW, Zheng XL, Tang CK. Apolipoprotein A-1 binding protein promotes macrophage cholesterol efflux by facilitating apolipoprotein A-1 binding to ABCA1 and preventing ABCA1 degradation. Atherosclerosis 2016; 248:149-59. [DOI: 10.1016/j.atherosclerosis.2016.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 01/27/2023]
|
27
|
The impact of structural genomics: the first quindecennial. ACTA ACUST UNITED AC 2016; 17:1-16. [PMID: 26935210 DOI: 10.1007/s10969-016-9201-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
The period 2000-2015 brought the advent of high-throughput approaches to protein structure determination. With the overall funding on the order of $2 billion (in 2010 dollars), the structural genomics (SG) consortia established worldwide have developed pipelines for target selection, protein production, sample preparation, crystallization, and structure determination by X-ray crystallography and NMR. These efforts resulted in the determination of over 13,500 protein structures, mostly from unique protein families, and increased the structural coverage of the expanding protein universe. SG programs contributed over 4400 publications to the scientific literature. The NIH-funded Protein Structure Initiatives alone have produced over 2000 scientific publications, which to date have attracted more than 93,000 citations. Software and database developments that were necessary to handle high-throughput structure determination workflows have led to structures of better quality and improved integrity of the associated data. Organized and accessible data have a positive impact on the reproducibility of scientific experiments. Most of the experimental data generated by the SG centers are freely available to the community and has been utilized by scientists in various fields of research. SG projects have created, improved, streamlined, and validated many protocols for protein production and crystallization, data collection, and functional analysis, significantly benefiting biological and biomedical research.
Collapse
|
28
|
Zheng H, Handing KB, Zimmerman MD, Shabalin IG, Almo SC, Minor W. X-ray crystallography over the past decade for novel drug discovery - where are we heading next? Expert Opin Drug Discov 2015; 10:975-89. [PMID: 26177814 DOI: 10.1517/17460441.2015.1061991] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Macromolecular X-ray crystallography has been the primary methodology for determining the three-dimensional structures of proteins, nucleic acids and viruses. Structural information has paved the way for structure-guided drug discovery and laid the foundations for structural bioinformatics. However, X-ray crystallography still has a few fundamental limitations, some of which may be overcome and complemented using emerging methods and technologies in other areas of structural biology. AREAS COVERED This review describes how structural knowledge gained from X-ray crystallography has been used to advance other biophysical methods for structure determination (and vice versa). This article also covers current practices for integrating data generated by other biochemical and biophysical methods with those obtained from X-ray crystallography. Finally, the authors articulate their vision about how a combination of structural and biochemical/biophysical methods may improve our understanding of biological processes and interactions. EXPERT OPINION X-ray crystallography has been, and will continue to serve as, the central source of experimental structural biology data used in the discovery of new drugs. However, other structural biology techniques are useful not only to overcome the major limitation of X-ray crystallography, but also to provide complementary structural data that is useful in drug discovery. The use of recent advancements in biochemical, spectroscopy and bioinformatics methods may revolutionize drug discovery, albeit only when these data are combined and analyzed with effective data management systems. Accurate and complete data management is crucial for developing experimental procedures that are robust and reproducible.
Collapse
Affiliation(s)
- Heping Zheng
- University of Virginia, Department of Molecular Physiology and Biological Physics , 1340 Jefferson Park Avenue, Charlottesville, VA 22908 , USA +1 434 243 6865 ; +1 434 243 2981 ;
| | | | | | | | | | | |
Collapse
|
29
|
ARIFIN GIRI-RACHMAN ERNAWATI, PRATAMA FENRYCO, ROKA AJI OKTIRA, PATRIATI ARUM, IHSANAWATI IHSANAWATI, RAMDANI MOEIS MAELITA, GIRI-RACHMAN PUTRA EDY, RAMDANI MOEIS MAELITA. Expression and Purification of PhoR Sensor-Domain Histidine Kinase of Mycobacterium tuberculosis in Eschericia coli. MICROBIOLOGY INDONESIA 2015. [DOI: 10.5454/mi.9.2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
30
|
NAD(P)H-hydrate dehydratase- a metabolic repair enzyme and its role in Bacillus subtilis stress adaptation. PLoS One 2014; 9:e112590. [PMID: 25393291 PMCID: PMC4231035 DOI: 10.1371/journal.pone.0112590] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/10/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND One of the strategies for survival stress conditions in bacteria is a regulatory adaptive system called general stress response (GSR), which is dependent on the SigB transcription factor in Bacillus sp. The GSR is one of the largest regulon in Bacillus sp., including about 100 genes; however, most of the genes that show changes in expression during various stresses have not yet been characterized or assigned a biochemical function for the encoded proteins. Previously, we characterized the Bacillus subtilis168 osmosensitive mutant, defective in the yxkO gene (encoding a putative ribokinase), which was recently assigned in vitro as an ADP/ATP-dependent NAD(P)H-hydrate dehydratase and was demonstrated to belong to the SigB operon. METHODS AND RESULTS We show the impact of YxkO on the activity of SigB-dependent Pctc promoter and adaptation to osmotic and ethanol stress and potassium limitation respectively. Using a 2DE approach, we compare the proteomes of WT and mutant strains grown under conditions of osmotic and ethanol stress. Both stresses led to changes in the protein level of enzymes that are involved in motility (flagellin), citrate cycle (isocitrate dehydrogenase, malate dehydrogenase), glycolysis (phosphoglycerate kinase), and decomposition of Amadori products (fructosamine-6-phosphate deglycase). Glutamine synthetase revealed a different pattern after osmotic stress. The patterns of enzymes for branched amino acid metabolism and cell wall synthesis (L-alanine dehydrogenase, aspartate-semialdehyde dehydrogenase, ketol-acid reductoisomerase) were altered after ethanol stress. CONCLUSION We performed the first characterization of a Bacillus subtilis168 knock-out mutant in the yxkO gene that encodes a metabolite repair enzyme. We show that such enzymes could play a significant role in the survival of stressed cells.
Collapse
|
31
|
Boyko KM, Gorbacheva MA, Rakitina TV, Korzhenevsky DA, Dorovatovsky PV, Lipkin AV, Popov VO. Identification of the ligand in the structure of the protein with unknown function STM4435 from Salmonella typhimurium. DOKL BIOCHEM BIOPHYS 2014; 457:121-4. [PMID: 25172330 DOI: 10.1134/s1607672914040012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Indexed: 01/09/2023]
Abstract
The unidentified ligand, which is present in the crystal of the protein with unknown function STM4435 from Salmonella typhimurium, was identified using a combination of high-resolution X-ray crystallography and accurate-mass time-of-flight mass spectrometry. The identified glycerol was present as a component of the solutions used for the isolation and crystallization of the protein and serves as the ligand mimicking the natural metabolite, presumably, 2-keto-myo-isonitol, which is indicative of the involvement of STM4435 in the myo-isonitol catabolism. The results of the present study show that this approach holds promise in complex studies aimed at determining, refining, or confirming the protein functions.
Collapse
Affiliation(s)
- K M Boyko
- Bach Institute of Biochemistry, Russian Academy of Sciences, Leninskii pr. 33, Moscow, 119071, Russia,
| | | | | | | | | | | | | |
Collapse
|
32
|
Advantages of crystallographic fragment screening: functional and mechanistic insights from a powerful platform for efficient drug discovery. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 116:92-100. [PMID: 25117499 DOI: 10.1016/j.pbiomolbio.2014.08.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 07/23/2014] [Accepted: 08/02/2014] [Indexed: 11/23/2022]
Abstract
X-ray crystallography has been an under-appreciated screening tool for fragment-based drug discovery due to the perception of low throughput and technical difficulty. Investigators in industry and academia have overcome these challenges by taking advantage of key factors that contribute to a successful crystallographic screening campaign. Efficient cocktail design and soaking methodologies have evolved to maximize throughput while minimizing false positives/negatives. In addition, technical improvements at synchrotron beamlines have dramatically increased data collection rates thus enabling screening on a timescale comparable to other techniques. The combination of available resources and efficient experimental design has resulted in many successful crystallographic screening campaigns. The three-dimensional crystal structure of the bound fragment complexed to its target, a direct result of the screening effort, enables structure-based drug design while revealing insights regarding protein dynamics and function not readily obtained through other experimental approaches. Furthermore, this "chemical interrogation" of the target protein crystals can lead to the identification of useful reagents for improving diffraction resolution or compound solubility.
Collapse
|
33
|
Carolan CG, Lamzin VS. Automated identification of crystallographic ligands using sparse-density representations. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:1844-53. [PMID: 25004962 PMCID: PMC4089483 DOI: 10.1107/s1399004714008578] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 04/15/2014] [Indexed: 12/05/2022]
Abstract
A novel procedure for the automatic identification of ligands in macromolecular crystallographic electron-density maps is introduced. It is based on the sparse parameterization of density clusters and the matching of the pseudo-atomic grids thus created to conformationally variant ligands using mathematical descriptors of molecular shape, size and topology. In large-scale tests on experimental data derived from the Protein Data Bank, the procedure could quickly identify the deposited ligand within the top-ranked compounds from a database of candidates. This indicates the suitability of the method for the identification of binding entities in fragment-based drug screening and in model completion in macromolecular structure determination.
Collapse
Affiliation(s)
- C. G. Carolan
- European Molecular Biology Laboratory (EMBL), c/o DESY, Notkestrasse 85, 22603 Hamburg, Germany
| | - V. S. Lamzin
- European Molecular Biology Laboratory (EMBL), c/o DESY, Notkestrasse 85, 22603 Hamburg, Germany
| |
Collapse
|
34
|
Occurrence and subcellular distribution of the NADPHX repair system in mammals. Biochem J 2014; 460:49-58. [PMID: 24611804 DOI: 10.1042/bj20131482] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hydration of NAD(P)H to NAD(P)HX, which inhibits several dehydrogenases, is corrected by an ATP-dependent dehydratase and an epimerase recently identified as the products of the vertebrate Carkd (carbohydrate kinase domain) and Aibp (apolipoprotein AI-binding protein) genes respectively. The purpose of the present study was to assess the presence of these enzymes in mammalian tissues and determine their subcellular localization. The Carkd gene encodes proteins with a predicted mitochondrial propeptide (mCARKD), a signal peptide (spCARKD) or neither of them (cCARKD). Confocal microscopy analysis of transfected CHO (Chinese-hamster ovary) cells indicated that cCARKD remains in the cytosol, whereas mCARKD and spCARKD are targeted to the mitochondria and the endoplasmic reticulum respectively. Unlike the other two forms, spCARKD is N-glycosylated, supporting its targeting to the endoplasmic reticulum. The Aibp gene encodes two different proteins, which we show to be targeted to the mitochondria (mAIBP) and the cytosol (cAIBP). Quantification of the NAD(P)HX dehydratase and epimerase activities in rat tissues, performed after partial purification, indicated that both enzymes are widely distributed, with total activities of ≈3-10 nmol/min per g of tissue. Liver fractionation by differential centrifugation confirmed the presence of the dehydratase and the epimerase in the cytosol and in mitochondria. These data support the notion that NAD(P)HX repair is extremely widespread.
Collapse
|
35
|
Edc3 function in yeast and mammals is modulated by interaction with NAD-related compounds. G3-GENES GENOMES GENETICS 2014; 4:613-22. [PMID: 24504254 PMCID: PMC4059234 DOI: 10.1534/g3.114.010470] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The control of mRNA translation and degradation is mediated in part by a set of proteins that can inhibit translation and promote decapping, as well as function in the assembly of cytoplasmic mRNP granules referred to as processing bodies (P-bodies). The conserved enhancer of mRNA decapping 3 (Edc3) protein functions to promote both decapping and P-body assembly. Crystal structures of the YjeF_N domain in hEdc3 identified a putative binding site for a small molecule. Structure modeling of the human Edc3 Yjef_N along with other Yjef_N-containing proteins suggests that this molecule is related to NAD(H). We now show human Edc3 directly binds NADH. We also show that human and yeast Edc3 chemically modify NAD in vitro. Mutations that are predicted to disrupt the binding and/or hydrolysis of an NAD-related molecule by yeast and human Edc3 affect the control of mRNA degradation and/or P-body composition in vivo. This suggests that the interaction of Edc3 with an NAD-related molecule affects its function in the regulation of mRNA translation and degradation and provides a possible mechanism to couple the energetics of the cell to posttranscriptional control. Moreover, this provides a unique example of and lends strength to the postulated connection of metabolites, enzymes, and RNA.
Collapse
|
36
|
Colinas M, Shaw HV, Loubéry S, Kaufmann M, Moulin M, Fitzpatrick TB. A pathway for repair of NAD(P)H in plants. J Biol Chem 2014; 289:14692-706. [PMID: 24706747 DOI: 10.1074/jbc.m114.556092] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Unwanted enzyme side reactions and spontaneous decomposition of metabolites can lead to a build-up of compounds that compete with natural enzyme substrates and must be dealt with for efficient metabolism. It has recently been realized that there are enzymes that process such compounds, formulating the concept of metabolite repair. NADH and NADPH are vital cellular redox cofactors but can form non-functional hydrates (named NAD(P)HX) spontaneously or enzymatically that compete with enzymes dependent on NAD(P)H, impairing normal enzyme function. Here we report on the functional characterization of components of a potential NAD(P)H repair pathway in plants comprising a stereospecific dehydratase (NNRD) and an epimerase (NNRE), the latter being fused to a vitamin B6 salvage enzyme. Through the use of the recombinant proteins, we show that the ATP-dependent NNRD and NNRE act concomitantly to restore NAD(P)HX to NAD(P)H. NNRD behaves as a tetramer and NNRE as a dimer, but the proteins do not physically interact. In vivo fluorescence analysis demonstrates that the proteins are localized to mitochondria and/or plastids, implicating these as the key organelles where this repair is required. Expression analysis indicates that whereas NNRE is present ubiquitously, NNRD is restricted to seeds but appears to be dispensable during the normal Arabidopsis life cycle.
Collapse
Affiliation(s)
- Maite Colinas
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Holly V Shaw
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Sylvain Loubéry
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Markus Kaufmann
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Michael Moulin
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Teresa B Fitzpatrick
- From the Department of Botany and Plant Biology, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
37
|
Fang L, Liu C, Miller YI. Zebrafish models of dyslipidemia: relevance to atherosclerosis and angiogenesis. Transl Res 2014; 163:99-108. [PMID: 24095954 PMCID: PMC3946603 DOI: 10.1016/j.trsl.2013.09.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/07/2013] [Accepted: 09/10/2013] [Indexed: 01/07/2023]
Abstract
Lipid and lipoprotein metabolism in zebrafish and in humans are remarkably similar. Zebrafish express all major nuclear receptors, lipid transporters, apolipoproteins and enzymes involved in lipoprotein metabolism. Unlike mice, zebrafish express cetp and the Cetp activity is detected in zebrafish plasma. Feeding zebrafish a high cholesterol diet, without any genetic intervention, results in significant hypercholesterolemia and robust lipoprotein oxidation, making zebrafish an attractive animal model to study mechanisms relevant to early development of human atherosclerosis. These studies are facilitated by the optical transparency of zebrafish larvae and the availability of transgenic zebrafish expressing fluorescent proteins in endothelial cells and macrophages. Thus, vascular processes can be monitored in live animals. In this review article, we discuss recent advances in using dyslipidemic zebrafish in atherosclerosis-related studies. We also summarize recent work connecting lipid metabolism with regulation of angiogenesis, the work that considerably benefited from using the zebrafish model. These studies uncovered the role of aibp, abca1, abcg1, mtp, apoB, and apoC2 in regulation of angiogenesis in zebrafish and paved the way for future studies in mammals, which may suggest new therapeutic approaches to modulation of excessive or diminished angiogenesis contributing to the pathogenesis of human disease.
Collapse
Affiliation(s)
- Longhou Fang
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Chao Liu
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
38
|
Abstract
INTRODUCTION X-ray crystallography plays an important role in structure-based drug design (SBDD), and accurate analysis of crystal structures of target macromolecules and macromolecule-ligand complexes is critical at all stages. However, whereas there has been significant progress in improving methods of structural biology, particularly in X-ray crystallography, corresponding progress in the development of computational methods (such as in silico high-throughput screening) is still on the horizon. Crystal structures can be overinterpreted and thus bias hypotheses and follow-up experiments. As in any experimental science, the models of macromolecular structures derived from X-ray diffraction data have their limitations, which need to be critically evaluated and well understood for structure-based drug discovery. AREAS COVERED This review describes how the validity, accuracy and precision of a protein or nucleic acid structure determined by X-ray crystallography can be evaluated from three different perspectives: i) the nature of the diffraction experiment; ii) the interpretation of an electron density map; and iii) the interpretation of the structural model in terms of function and mechanism. The strategies to optimally exploit a macromolecular structure are also discussed in the context of 'Big Data' analysis, biochemical experimental design and structure-based drug discovery. EXPERT OPINION Although X-ray crystallography is one of the most detailed 'microscopes' available today for examining macromolecular structures, the authors would like to re-emphasize that such structures are only simplified models of the target macromolecules. The authors also wish to reinforce the idea that a structure should not be thought of as a set of precise coordinates but rather as a framework for generating hypotheses to be explored. Numerous biochemical and biophysical experiments, including new diffraction experiments, can and should be performed to verify or falsify these hypotheses. X-ray crystallography will find its future application in drug discovery by the development of specific tools that would allow realistic interpretation of the outcome coordinates and/or support testing of these hypotheses.
Collapse
Affiliation(s)
- Heping Zheng
- University of Virginia, Department of Molecular Physiology and Biological Physics, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
- Center for Structural Genomics of Infectious Diseases (CSGID)
- Midwest Center for Structural Genomics (MCSG), USA
- New York Structural Genomics Research Consortium (NYSGRC), USA
- Specializes in Protein Crystallography, Data Analytics and Data Mining, Research Scientist
| | - Jing Hou
- University of Virginia, Department of Molecular Physiology and Biological Physics, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
- Center for Structural Genomics of Infectious Diseases (CSGID)
- Enzyme Structure Initiative (EFI), USA
- New York Structural Genomics Research Consortium (NYSGRC), USA
- Specializes in Protein Crystallography, Research Associate
| | - Matthew D Zimmerman
- University of Virginia, Department of Molecular Physiology and Biological Physics, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
- Center for Structural Genomics of Infectious Diseases (CSGID)
- Enzyme Structure Initiative (EFI), USA
- Midwest Center for Structural Genomics (MCSG), USA
- New York Structural Genomics Research Consortium (NYSGRC), USA
- Specializes in Protein Crystallography, Data Mining and Management, Instructor of Research
| | - Alexander Wlodawer
- National Cancer Institute, Center for Cancer Research, Frederick, MD 21702, USA
- Specializes in Macromolecular Structure and Function, Chief of the Macromolecular Crystallography Laboratory
| | - Wladek Minor
- University of Virginia, Department of Molecular Physiology and Biological Physics, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
- Center for Structural Genomics of Infectious Diseases (CSGID)
- Enzyme Structure Initiative (EFI), USA
- Midwest Center for Structural Genomics (MCSG), USA
- New York Structural Genomics Research Consortium (NYSGRC), USA
- Specializes in Structural Biology, Data Mining and Management, Professor
| |
Collapse
|
39
|
Wlodawer A, Minor W, Dauter Z, Jaskolski M. Protein crystallography for aspiring crystallographers or how to avoid pitfalls and traps in macromolecular structure determination. FEBS J 2013; 280:5705-36. [PMID: 24034303 DOI: 10.1111/febs.12495] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/12/2013] [Accepted: 08/20/2013] [Indexed: 12/28/2022]
Abstract
The number of macromolecular structures deposited in the Protein Data Bank now approaches 100,000, with the vast majority of them determined by crystallographic methods. Thousands of papers describing such structures have been published in the scientific literature, and 20 Nobel Prizes in chemistry or medicine have been awarded for discoveries based on macromolecular crystallography. New hardware and software tools have made crystallography appear to be an almost routine (but still far from being analytical) technique and many structures are now being determined by scientists with very limited experience in the practical aspects of the field. However, this apparent ease is sometimes illusory and proper procedures need to be followed to maintain high standards of structure quality. In addition, many noncrystallographers may have problems with the critical evaluation and interpretation of structural results published in the scientific literature. The present review provides an outline of the technical aspects of crystallography for less experienced practitioners, as well as information that might be useful for users of macromolecular structures, aiming to show them how to interpret (but not overinterpret) the information present in the coordinate files and in their description. A discussion of the extent of information that can be gleaned from the atomic coordinates of structures solved at different resolution is provided, as well as problems and pitfalls encountered in structure determination and interpretation.
Collapse
Affiliation(s)
- Alexander Wlodawer
- Protein Structure Section, Macromolecular Crystallography Laboratory, NCI at Frederick, Frederick, MD, USA
| | | | | | | |
Collapse
|
40
|
Majorek KA, Kuhn ML, Chruszcz M, Anderson WF, Minor W. Structural, functional, and inhibition studies of a Gcn5-related N-acetyltransferase (GNAT) superfamily protein PA4794: a new C-terminal lysine protein acetyltransferase from pseudomonas aeruginosa. J Biol Chem 2013; 288:30223-30235. [PMID: 24003232 DOI: 10.1074/jbc.m113.501353] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Gcn5-related N-acetyltransferase (GNAT) superfamily is a large group of evolutionarily related acetyltransferases, with multiple paralogs in organisms from all kingdoms of life. The functionally characterized GNATs have been shown to catalyze the transfer of an acetyl group from acetyl-coenzyme A (Ac-CoA) to the amine of a wide range of substrates, including small molecules and proteins. GNATs are prevalent and implicated in a myriad of aspects of eukaryotic and prokaryotic physiology, but functions of many GNATs remain unknown. In this work, we used a multi-pronged approach of x-ray crystallography and biochemical characterization to elucidate the sequence-structure-function relationship of the GNAT superfamily member PA4794 from Pseudomonas aeruginosa. We determined that PA4794 acetylates the Nε amine of a C-terminal lysine residue of a peptide, suggesting it is a protein acetyltransferase specific for a C-terminal lysine of a substrate protein or proteins. Furthermore, we identified a number of molecules, including cephalosporin antibiotics, which are inhibitors of PA4794 and bind in its substrate-binding site. Often, these molecules mimic the conformation of the acetylated peptide product. We have determined structures of PA4794 in the apo-form, in complexes with Ac-CoA, CoA, several antibiotics and other small molecules, and a ternary complex with the products of the reaction: CoA and acetylated peptide. Also, we analyzed PA4794 mutants to identify residues important for substrate binding and catalysis.
Collapse
Affiliation(s)
- Karolina A Majorek
- From the Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908,; the Bioinformatics Laboratory, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland,; the Midwest Center for Structural Genomics, Argonne National Laboratory, Argonne, Illinois 60439, and; the Center for Structural Genomics of Infectious Diseases (CSGID)
| | - Misty L Kuhn
- the Center for Structural Genomics of Infectious Diseases (CSGID); the Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Maksymilian Chruszcz
- From the Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908,; the Midwest Center for Structural Genomics, Argonne National Laboratory, Argonne, Illinois 60439, and; the Center for Structural Genomics of Infectious Diseases (CSGID); the Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208
| | - Wayne F Anderson
- the Midwest Center for Structural Genomics, Argonne National Laboratory, Argonne, Illinois 60439, and; the Center for Structural Genomics of Infectious Diseases (CSGID); the Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Wladek Minor
- From the Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908,; the Midwest Center for Structural Genomics, Argonne National Laboratory, Argonne, Illinois 60439, and; the Center for Structural Genomics of Infectious Diseases (CSGID).
| |
Collapse
|
41
|
Van Schaftingen E, Rzem R, Marbaix A, Collard F, Veiga-da-Cunha M, Linster CL. Metabolite proofreading, a neglected aspect of intermediary metabolism. J Inherit Metab Dis 2013; 36:427-34. [PMID: 23296366 DOI: 10.1007/s10545-012-9571-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 11/26/2012] [Accepted: 11/29/2012] [Indexed: 10/27/2022]
Abstract
Enzymes of intermediary metabolism are less specific than what is usually assumed: they often act on metabolites that are not their 'true' substrate, making abnormal metabolites that may be deleterious if they accumulate. Some of these abnormal metabolites are reconverted to normal metabolites by repair enzymes, which play therefore a role akin to the proofreading activities of DNA polymerases and aminoacyl-tRNA synthetases. An illustrative example of such repair enzymes is L-2-hydroxyglutarate dehydrogenase, which eliminates a metabolite abnormally made by a Krebs cycle enzyme. Mutations in L-2-hydroxyglutarate dehydrogenase lead to L-2-hydroxyglutaric aciduria, a leukoencephalopathy. Other examples are the epimerase and the ATP-dependent dehydratase that repair hydrated forms of NADH and NADPH; ethylmalonyl-CoA decarboxylase, which eliminates an abnormal metabolite formed by acetyl-CoA carboxylase, an enzyme of fatty acid synthesis; L-pipecolate oxidase, which repairs a metabolite formed by a side activity of an enzyme of L-proline biosynthesis. Metabolite proofreading enzymes are likely quite common, but most of them are still unidentified. A defect in these enzymes may account for new metabolic disorders.
Collapse
|
42
|
Abstract
In this issue of Structure, Shumilin and colleagues show the power of metabolite screening by means of X-ray crystallography to link an orphan protein domain with an orphan biochemical function. This result paves the way for large-scale functional annotation and sets new objectives for structural genomics.
Collapse
|