Generic placeholder image

Current Drug Metabolism

Editor-in-Chief

ISSN (Print): 1389-2002
ISSN (Online): 1875-5453

Mini-Review Article

State-of-the-art Application of Artificial Intelligence to Transporter-centered Functional and Pharmaceutical Research

Author(s): Jiayi Yin, Nanxin You, Fengcheng Li, Mingkun Lu, Su Zeng* and Feng Zhu*

Volume 24, Issue 3, 2023

Published on: 08 June, 2023

Page: [162 - 174] Pages: 13

DOI: 10.2174/1389200224666230523155759

Price: $65

conference banner
Abstract

Protein transporters not only have essential functions in regulating the transport of endogenous substrates and remote communication between organs and organisms, but they also play a vital role in drug absorption, distribution, and excretion and are recognized as major determinants of drug safety and efficacy. Understanding transporter function is important for drug development and clarifying disease mechanisms. However, the experimental-based functional research on transporters has been challenged and hinged by the expensive cost of time and resources. With the increasing volume of relevant omics datasets and the rapid evolution of artificial intelligence (AI) techniques, next-generation AI is becoming increasingly prevalent in the functional and pharmaceutical research of transporters. Thus, a comprehensive discussion on the state-of-the-art application of AI in three cutting-edge directions was provided in this review, which included (a) transporter classification and function annotation, (b) structure discovery of membrane transporters, and (c) drug-transporter interaction prediction. This study provides a panoramic view of AI algorithms and tools applied to the field of transporters. It is expected to guide a better understanding and utilization of AI techniques for in-depth studies of transporter-centered functional and pharmaceutical research.

Graphical Abstract

[1]
Nigam, S.K. What do drug transporters really do? Nat. Rev. Drug Discov., 2015, 14(1), 29-44.
[http://dx.doi.org/10.1038/nrd4461] [PMID: 25475361]
[2]
Jiang, T.; Wen, P.C.; Trebesch, N.; Zhao, Z.; Pant, S.; Kapoor, K.; Shekhar, M.; Tajkhorshid, E. Computational dissection of membrane transport at a microscopic level. Trends Biochem. Sci., 2020, 45(3), 202-216.
[http://dx.doi.org/10.1016/j.tibs.2019.09.001] [PMID: 31813734]
[3]
Song, W.; Li, D.; Tao, L.; Luo, Q.; Chen, L. Solute carrier transporters: The metabolic gatekeepers of immune cells. Acta Pharm. Sin. B, 2020, 10(1), 61-78.
[http://dx.doi.org/10.1016/j.apsb.2019.12.006] [PMID: 31993307]
[4]
César-Razquin, A.; Snijder, B.; Frappier-Brinton, T.; Isserlin, R.; Gyimesi, G.; Bai, X.; Reithmeier, R.A.; Hepworth, D.; Hediger, M.A.; Edwards, A.M.; Superti-Furga, G. A call for systematic research on solute carriers. Cell, 2015, 162(3), 478-487.
[http://dx.doi.org/10.1016/j.cell.2015.07.022] [PMID: 26232220]
[5]
Czuba, L.C.; Hillgren, K.M.; Swaan, P.W. Post-translational modifications of transporters. Pharmacol. Ther., 2018, 192(1), 88-99.
[http://dx.doi.org/10.1016/j.pharmthera.2018.06.013] [PMID: 29966598]
[6]
Giacomini, K.M.; Huang, S.M.; Tweedie, D.J.; Benet, L.Z.; Brouwer, K.L.R.; Chu, X.; Dahlin, A.; Evers, R.; Fischer, V.; Hillgren, K.M.; Hoffmaster, K.A.; Ishikawa, T.; Keppler, D.; Kim, R.B.; Lee, C.A.; Niemi, M.; Polli, J.W.; Sugiyama, Y.; Swaan, P.W.; Ware, J.A.; Wright, S.H.; Wah Yee, S.; Zamek-Gliszczynski, M.J.; Zhang, L. Membrane transporters in drug development. Nat. Rev. Drug Discov., 2010, 9(3), 215-236.
[http://dx.doi.org/10.1038/nrd3028] [PMID: 20190787]
[7]
Fung, S.W.; Cheung, P.F.Y.; Yip, C.W.; Ng, L.W.C.; Cheung, T.T.; Chong, C.C.N.; Lee, C.; Lai, P.B.S.; Chan, A.W.H.; Tsao, G.S.W.; Wong, C.H.; Chan, S.L.; Lo, K.W.; Cheung, S.T. The ATP-binding cassette transporter ABCF1 is a hepatic oncofetal protein that promotes chemoresistance, EMT and cancer stemness in hepatocellular carcinoma. Cancer Lett., 2019, 457(1), 98-109.
[http://dx.doi.org/10.1016/j.canlet.2019.05.010] [PMID: 31100412]
[8]
Lin, L.; Yee, S.W.; Kim, R.B.; Giacomini, K.M. SLC transporters as therapeutic targets: Emerging opportunities. Nat. Rev. Drug Discov., 2015, 14(8), 543-560.
[http://dx.doi.org/10.1038/nrd4626] [PMID: 26111766]
[9]
Yang, M.; Ma, J.; Ruan, J.; Ye, Y.; Fu, P.P.C.; Lin, G. Intestinal and hepatic biotransformation of pyrrolizidine alkaloid N-oxides to toxic pyrrolizidine alkaloids. Arch. Toxicol., 2019, 93(8), 2197-2209.
[http://dx.doi.org/10.1007/s00204-019-02499-2] [PMID: 31222523]
[10]
König, J.; Müller, F.; Fromm, M.F. Transporters and drug-drug interactions: Important determinants of drug disposition and effects. Pharmacol. Rev., 2013, 65(3), 944-966.
[http://dx.doi.org/10.1124/pr.113.007518] [PMID: 23686349]
[11]
Wang, J.Q.; Yang, Y.; Cai, C.Y.; Teng, Q.X.; Cui, Q.; Lin, J. Multidrug resistance proteins (MRPs): Structure, function and the overcoming of cancer multidrug resistance. Drug Resist. Updat., 2021, 54, 100743.
[12]
Kim, J.; Tan, Y.Z.; Wicht, K.J.; Erramilli, S.K.; Dhingra, S.K.; Okombo, J.; Vendome, J.; Hagenah, L.M.; Giacometti, S.I.; Warren, A.L.; Nosol, K.; Roepe, P.D.; Potter, C.S.; Carragher, B.; Kossiakoff, A.A.; Quick, M.; Fidock, D.A.; Mancia, F. Structure and drug resistance of the Plasmodium falciparum transporter PfCRT. Nature, 2019, 576(7786), 315-320.
[http://dx.doi.org/10.1038/s41586-019-1795-x] [PMID: 31776516]
[13]
Harris, A.; Wagner, M.; Du, D.; Raschka, S.; Nentwig, L.M.; Gohlke, H.; Smits, S.H.J.; Luisi, B.F.; Schmitt, L. Structure and efflux mechanism of the yeast pleiotropic drug resistance transporter Pdr5. Nat. Commun., 2021, 12(1), 5254.
[http://dx.doi.org/10.1038/s41467-021-25574-8] [PMID: 34489436]
[14]
Alam, A.; Kowal, J.; Broude, E.; Roninson, I.; Locher, K.P. Structural insight into substrate and inhibitor discrimination by human P-glycoprotein. Science, 2019, 363(6428), 753-756.
[http://dx.doi.org/10.1126/science.aav7102] [PMID: 30765569]
[15]
Montanari, F.; Ecker, G.F. Prediction of drug–ABC-transporter interaction — Recent advances and future challenges. Adv. Drug Deliv. Rev., 2015, 86(1), 17-26.
[http://dx.doi.org/10.1016/j.addr.2015.03.001] [PMID: 25769815]
[16]
Trueck, C.; Hsin, C.; Scherf-Clavel, O.; Schaeffeler, E.; Lenssen, R.; Gazzaz, M.; Gersie, M.; Taubert, M.; Quasdorff, M.; Schwab, M.; Kinzig, M.; Sörgel, F.; Stoffel, M.S.; Fuhr, U. A clinical drug-drug interaction study assessing a novel drug transporter phenotyping cocktail with adefovir, sitagliptin, metformin, pitavastatin, and digoxin. Clin. Pharmacol. Ther., 2019, 106(6), 1398-1407.
[http://dx.doi.org/10.1002/cpt.1564] [PMID: 31247117]
[17]
Chu, X.; Liao, M.; Shen, H.; Yoshida, K.; Zur, A.A.; Arya, V.; Galetin, A.; Giacomini, K.M.; Hanna, I.; Kusuhara, H.; Lai, Y.; Rodrigues, D.; Sugiyama, Y.; Zamek-Gliszczynski, M.J.; Zhang, L. Clinical probes and endogenous biomarkers as substrates for transporter drug-drug interaction evaluation: Perspectives from the international transporter consortium. Clin. Pharmacol. Ther., 2018, 104(5), 836-864.
[http://dx.doi.org/10.1002/cpt.1216] [PMID: 30347454]
[18]
David, R.; Byrt, C.S.; Tyerman, S.D.; Gilliham, M.; Wege, S. Roles of membrane transporters: Connecting the dots from sequence to phenotype. Ann. Bot., 2019, 124(2), 201-208.
[http://dx.doi.org/10.1093/aob/mcz066] [PMID: 31162525]
[19]
Garibsingh, R.A.A.; Schlessinger, A. Advances and challenges in rational drug design for SLCs. Trends Pharmacol. Sci., 2019, 40(10), 790-800.
[http://dx.doi.org/10.1016/j.tips.2019.08.006] [PMID: 31519459]
[20]
Lee, S.C.; Arya, V.; Yang, X.; Volpe, D.A.; Zhang, L. Evaluation of transporters in drug development: Current status and contemporary issues. Adv. Drug Deliv. Rev., 2017, 116(1), 100-118.
[http://dx.doi.org/10.1016/j.addr.2017.07.020] [PMID: 28760687]
[21]
Basile, A.O.; Yahi, A.; Tatonetti, N.P. Artificial intelligence for drug toxicity and safety. Trends Pharmacol. Sci., 2019, 40(9), 624-635.
[http://dx.doi.org/10.1016/j.tips.2019.07.005] [PMID: 31383376]
[22]
Zhu, C.; Cai, T.; Jin, Y.; Chen, J.; Liu, G.; Xu, N.; Shen, R.; Chen, Y.; Han, L.; Wang, S.; Wu, C.; Zhu, M. Artificial intelligence and network pharmacology based investigation of pharmacological mechanism and substance basis of Xiaokewan in treating diabetes. Pharmacol. Res., 2020, 159(1), 104935.
[http://dx.doi.org/10.1016/j.phrs.2020.104935] [PMID: 32464328]
[23]
Fan, K.; Cheng, L.; Li, L. Artificial intelligence and machine learning methods in predicting anti-cancer drug combination effects. Brief. Bioinform., 2021, 22(6), bbab271.
[http://dx.doi.org/10.1093/bib/bbab271] [PMID: 34347041]
[24]
Yin, J.; Li, F.; Zhou, Y.; Mou, M.; Lu, Y.; Chen, K.; Xue, J.; Luo, Y.; Fu, J.; He, X.; Gao, J.; Zeng, S.; Yu, L.; Zhu, F. INTEDE: Interactome of drug-metabolizing enzymes. Nucleic Acids Res., 2021, 49(D1), D1233-D1243.
[http://dx.doi.org/10.1093/nar/gkaa755] [PMID: 33045737]
[25]
Yang, Q.; Wang, Y.; Zhang, Y.; Li, F.; Xia, W.; Zhou, Y.; Qiu, Y.; Li, H.; Zhu, F. NOREVA: Enhanced normalization and evaluation of time-course and multi-class metabolomic data. Nucleic Acids Res., 2020, 48(W1), W436-W448.
[http://dx.doi.org/10.1093/nar/gkaa258] [PMID: 32324219]
[26]
Tang, J.; Fu, J.; Wang, Y.; Li, B.; Li, Y.; Yang, Q.; Cui, X.; Hong, J.; Li, X.; Chen, Y.; Xue, W.; Zhu, F. Anpela: Analysis and performance assessment of the label-free quantification workflow for metaproteomic studies. Brief. Bioinform., 2020, 21(2), 621-636.
[http://dx.doi.org/10.1093/bib/bby127] [PMID: 30649171]
[27]
Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S.A.A.; Ballard, A.J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A.W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly accurate protein structure prediction with AlphaFold. Nature, 2021, 596(7873), 583-589.
[http://dx.doi.org/10.1038/s41586-021-03819-2] [PMID: 34265844]
[28]
Mullard, A. What does AlphaFold mean for drug discovery? Nat. Rev. Drug Discov., 2021, 20(10), 725-727.
[http://dx.doi.org/10.1038/d41573-021-00161-0] [PMID: 34522032]
[29]
Cao, Y.; Shen, Y. TALE: Transformer-based protein function Annotation with joint sequence–Label Embedding. Bioinformatics, 2021, 37(18), 2825-2833.
[http://dx.doi.org/10.1093/bioinformatics/btab198] [PMID: 33755048]
[30]
Hong, J.; Luo, Y.; Zhang, Y.; Ying, J.; Xue, W.; Xie, T.; Tao, L.; Zhu, F. Protein functional annotation of simultaneously improved stability, accuracy and false discovery rate achieved by a sequence-based deep learning. Brief. Bioinform., 2020, 21(4), 1437-1447.
[http://dx.doi.org/10.1093/bib/bbz081] [PMID: 31504150]
[31]
Jendele, L.; Krivak, R.; Skoda, P.; Novotny, M.; Hoksza, D. PrankWeb: A web server for ligand binding site prediction and visualization. Nucleic Acids Res., 2019, 47(W1), W345-W349.
[http://dx.doi.org/10.1093/nar/gkz424] [PMID: 31114880]
[32]
Krivák, R.; Hoksza, D. P2Rank: machine learning based tool for rapid and accurate prediction of ligand binding sites from protein structure. J. Cheminform., 2018, 10(1), 39.
[http://dx.doi.org/10.1186/s13321-018-0285-8] [PMID: 30109435]
[33]
Iwabuchi, Y.; Kameyama, M.; Matsusaka, Y.; Narimatsu, H.; Hashimoto, M.; Seki, M.; Ito, D.; Tabuchi, H.; Yamada, Y.; Jinzaki, M. A diagnostic strategy for Parkinsonian syndromes using quantitative indices of DAT SPECT and MIBG scintigraphy: An investigation using the classification and regression tree analysis. Eur. J. Nucl. Med. Mol. Imag, 2021, 48(6), 1833-1841.
[http://dx.doi.org/10.1007/s00259-020-05168-0] [PMID: 33392714]
[34]
Nigam, A.K.; Ojha, A.A.; Li, J.G.; Shi, D.; Bhatnagar, V.; Nigam, K.B.; Abagyan, R.; Nigam, S.K. Molecular properties of drugs handled by kidney OATs and liver OATPs revealed by chemoinformatics and machine learning: Implications for kidney and liver disease. Pharmaceutics, 2021, 13(10), 1720.
[http://dx.doi.org/10.3390/pharmaceutics13101720] [PMID: 34684013]
[35]
Saier, M.H., Jr; Reddy, V.S.; Moreno-Hagelsieb, G.; Hendargo, K.J.; Zhang, Y.; Iddamsetty, V.; Lam, K.J.K.; Tian, N.; Russum, S.; Wang, J.; Medrano-Soto, A. The transporter classification database (TCDB): 2021 update. Nucleic Acids Res., 2021, 49(D1), D461-D467.
[http://dx.doi.org/10.1093/nar/gkaa1004] [PMID: 33170213]
[36]
Elbourne, L.D.H.; Tetu, S.G.; Hassan, K.A.; Paulsen, I.T. TransportDB 2.0: A database for exploring membrane transporters in sequenced genomes from all domains of life. Nucleic Acids Res., 2017, 45(D1), D320-D324.
[http://dx.doi.org/10.1093/nar/gkw1068] [PMID: 27899676]
[37]
Radi, M.S. SalcedoSora, J.E.; Kim, S.H.; Sudarsan, S.; Sastry, A.V.; Kell, D.B.; Herrgård, M.J.; Feist, A.M. Membrane transporter identification and modulation via adaptive laboratory evolution. Metab. Eng., 2022, 72, 376-390.
[http://dx.doi.org/10.1016/j.ymben.2022.05.004] [PMID: 35598887]
[38]
Alballa, M.; Butler, G. TooT-T: Discrimination of transport proteins from non-transport proteins. BMC Bioinformat, 2020, 21(S3), 25.
[http://dx.doi.org/10.1186/s12859-019-3311-6] [PMID: 32321420]
[39]
Potter, S.C.; Luciani, A.; Eddy, S.R.; Park, Y.; Lopez, R.; Finn, R.D. HMMER web server: 2018 update. Nucleic Acids Res., 2018, 46(W1), W200-W204.
[http://dx.doi.org/10.1093/nar/gky448] [PMID: 29905871]
[40]
Ebrahimie, E.; Zamansani, F.; Alanazi, I.O.; Sabi, E.M.; Khazandi, M.; Ebrahimi, F.; Mohammadi-Dehcheshmeh, M.; Ebrahimi, M. Advances in understanding the specificity function of transporters by machine learning. Comput. Biol. Med., 2021, 138(1), 104893.
[http://dx.doi.org/10.1016/j.compbiomed.2021.104893] [PMID: 34598069]
[41]
Tetko, I.V.; Rodchenkov, I.V.; Walter, M.C.; Rattei, T.; Mewes, H.W. Beyond the ‘best’ match: Machine learning annotation of protein sequences by integration of different sources of information. Bioinformatics, 2008, 24(5), 621-628.
[http://dx.doi.org/10.1093/bioinformatics/btm633] [PMID: 18174184]
[42]
Ashrafuzzaman, M. Artificial intelligence, machine learning and deep learning in ion channel bioinformatics. Membranes, 2021, 11(9), 672.
[http://dx.doi.org/10.3390/membranes11090672] [PMID: 34564489]
[43]
Wang, G.; Lu, J.; Choi, K.S.; Zhang, G. A transfer-based additive LS-SVM classifier for handling missing data. IEEE Trans. Cybern., 2020, 50(2), 739-752.
[http://dx.doi.org/10.1109/TCYB.2018.2872800] [PMID: 30334775]
[44]
Barash, E.; Sal-Man, N.; Sabato, S.; Ziv-Ukelson, M. BacPaCS-bacterial pathogenicity classification via sparse-SVM. Bioinformatics, 2019, 35(12), 2001-2008.
[http://dx.doi.org/10.1093/bioinformatics/bty928] [PMID: 30407484]
[45]
Huang, S.; Cai, N.; Pacheco, P.P.; Narrandes, S.; Wang, Y.; Xu, W. Applications of support vector machine (SVM) learning in cancer genomics. Cancer Genom Proteom, 2018, 15(1), 41-51.
[PMID: 29275361]
[46]
Gupta, R.; Srivastava, D.; Sahu, M.; Tiwari, S.; Ambasta, R.K.; Kumar, P. Artificial intelligence to deep learning: Machine intelligence approach for drug discovery. Mol. Divers., 2021, 25(3), 1315-1360.
[http://dx.doi.org/10.1007/s11030-021-10217-3] [PMID: 33844136]
[47]
Zhang, F.; Petersen, M.; Johnson, L.; Hall, J.; O’Bryant, S.E. Recursive support vector machine biomarker selection for Alzheimer’s disease. J. Alzheimers Dis., 2021, 79(4), 1691-1700.
[http://dx.doi.org/10.3233/JAD-201254] [PMID: 33492292]
[48]
Taju, S.W.; Shah, S.M.A.; Ou, Y.Y. ActTRANS: Functional classification in active transport proteins based on transfer learning and contextual representations. Comput. Biol. Chem., 2021, 93(1), 107537.
[http://dx.doi.org/10.1016/j.compbiolchem.2021.107537] [PMID: 34217007]
[49]
Liou, Y.F.; Vasylenko, T.; Yeh, C.L.; Lin, W.C.; Chiu, S.H.; Charoenkwan, P.; Shu, L.S.; Ho, S.Y.; Huang, H.L. SCMMTP: Identifying and characterizing membrane transport proteins using propensity scores of dipeptides. BMC Genomics, 2015, 16(S12), S6.
[http://dx.doi.org/10.1186/1471-2164-16-S12-S6] [PMID: 26677931]
[50]
Gligorijević V.; Barot, M.; Bonneau, R. deepNF: Deep network fusion for protein function prediction. Bioinformatics, 2018, 34(22), 3873-3881.
[http://dx.doi.org/10.1093/bioinformatics/bty440] [PMID: 29868758]
[51]
Asadi, S.; Roshan, S.; Kattan, M.W. Random forest swarm optimization-based for heart diseases diagnosis. J. Biomed. Inform., 2021, 115(1), 103690.
[http://dx.doi.org/10.1016/j.jbi.2021.103690] [PMID: 33540075]
[52]
Sarica, A.; Cerasa, A.; Quattrone, A. Random forest algorithm for the classification of neuroimaging data in Alzheimer’s disease: A systematic review. Front. Aging Neurosci., 2017, 9(1), 329.
[http://dx.doi.org/10.3389/fnagi.2017.00329] [PMID: 29056906]
[53]
Hussain, D.; Han, S.M.; Kim, T.S. Automatic hip geometric feature extraction in DXA imaging using regional random forest. J. XRay Sci. Technol., 2019, 27(2), 207-236.
[http://dx.doi.org/10.3233/XST-180434] [PMID: 30594942]
[54]
Sakiyama, Y. The use of machine learning and nonlinear statistical tools for ADME prediction. Expert Opin. Drug Metab. Toxicol., 2009, 5(2), 149-169.
[http://dx.doi.org/10.1517/17425250902753261] [PMID: 19239395]
[55]
Gajowniczek, K.; Grzegorczyk, I. Ząbkowski, T.; Bajaj, C. Weighted random forests to improve arrhythmia classification. Electronics , 2020, 9(1), 99.
[http://dx.doi.org/10.3390/electronics9010099] [PMID: 32051761]
[56]
Hanko, M.; Grendár, M.; Snopko, P.; Opšenák, R.; Šutovský, J. Benčo, M.; Soršák, J.; Zeleňák, K.; Kolarovszki, B. Random forest-based prediction of outcome and mortality in patients with traumatic brain injury undergoing primary decompressive craniectomy. World Neurosurg., 2021, 148(1), e450-e458.
[http://dx.doi.org/10.1016/j.wneu.2021.01.002] [PMID: 33444843]
[57]
Hou, R.; Wang, L.; Wu, Y.J. Predicting ATP-binding cassette transporters using the random forest method. Front. Genet., 2020, 11(1), 156.
[http://dx.doi.org/10.3389/fgene.2020.00156] [PMID: 32269586]
[58]
Ru, X.; Li, L.; Zou, Q. Incorporating distance-based top-n-gram and random forest to identify electron transport proteins. J. Proteome Res., 2019, 18(7), 2931-2939.
[http://dx.doi.org/10.1021/acs.jproteome.9b00250] [PMID: 31136183]
[59]
Khare, S.K.; Bajaj, V. Time-frequency representation and convolutional neural network-based emotion recognition. IEEE Trans. Neural Netw. Learn. Syst., 2021, 32(7), 2901-2909.
[http://dx.doi.org/10.1109/TNNLS.2020.3008938] [PMID: 32735536]
[60]
Liu, W.; Lu, B. Multi-stream convolutional neural network-based wearable, flexible bionic gesture surface muscle feature extraction and recognition. Front. Bioeng. Biotechnol., 2022, 10(1), 833793.
[http://dx.doi.org/10.3389/fbioe.2022.833793] [PMID: 35310001]
[61]
Soffer, S.; Ben-Cohen, A.; Shimon, O.; Amitai, M.M.; Greenspan, H.; Klang, E. Convolutional neural networks for radiologic images: A radiologist’s guide. Radiology, 2019, 290(3), 590-606.
[http://dx.doi.org/10.1148/radiol.2018180547] [PMID: 30694159]
[62]
Ho, Q.T.; Le, N.Q.K.; Ou, Y.Y. m CNN-ETC: Identifying electron transporters and their functional families by using multiple windows scanning techniques in convolutional neural networks with evolutionary information of protein sequences. Brief. Bioinform., 2022, 23(1), bbab352.
[http://dx.doi.org/10.1093/bib/bbab352] [PMID: 34472594]
[63]
Taju, S.W.; Ou, Y.Y. DeepIon: Deep learning approach for classifying ion transporters and ion channels from membrane proteins. J. Comput. Chem., 2019, 40(15), 1521-1529.
[http://dx.doi.org/10.1002/jcc.25805] [PMID: 30883833]
[64]
Zhang, D; Kabuka, MR Protein family classification from scratch: A CNN based deep learning approach. IEEE/ACM Transact. Computat.Biol. Bioinformat., 2021, 18(5)
[http://dx.doi.org/10.1109/TCBB.2020.2966633]
[65]
Taju, S.W.; Nguyen, T.T.D.; Le, N.Q.K.; Kusuma, R.M.I.; Ou, Y.Y. DeepEfflux: A 2D convolutional neural network model for identifying families of efflux proteins in transporters. Bioinformatics, 2018, 34(18), 3111-3117.
[http://dx.doi.org/10.1093/bioinformatics/bty302] [PMID: 29668844]
[66]
Nakayama, Y.; Mukai, N.; Kreitzer, G.; Patwari, P.; Yoshioka, J. Interaction of ARRDC4 with GLUT1 mediates metabolic stress in the ischemic heart. Circ. Res., 2022, 131(6), 510-527.
[http://dx.doi.org/10.1161/CIRCRESAHA.122.321351] [PMID: 35950500]
[67]
Mermer, F.; Poliquin, S.; Zhou, S.; Wang, X.; Ding, Y.; Yin, F.; Shen, W.; Wang, J.; Rigsby, K.; Xu, D.; Mack, T.; Nwosu, G.; Flamm, C.; Stein, M.; Kang, J.Q. Astrocytic GABA transporter 1 deficit in novel SLC6A1 variants mediated epilepsy: Connected from protein destabilization to seizures in mice and humans. Neurobiol. Dis., 2022, 172, 105810.
[http://dx.doi.org/10.1016/j.nbd.2022.105810] [PMID: 35840120]
[68]
Koleske, M.L.; McInnes, G.; Brown, J.E.H.; Thomas, N.; Hutchinson, K.; Chin, M.Y.; Koehl, A.; Arkin, M.R.; Schlessinger, A.; Gallagher, R.C.; Song, Y.S.; Altman, R.B.; Giacomini, K.M. Functional genomics of OCTN2 variants informs protein-specific variant effect predictor for Carnitine Transporter Deficiency. Proc. Natl. Acad. Sci. USA, 2022, 119(46), e2210247119.
[http://dx.doi.org/10.1073/pnas.2210247119] [PMID: 36343260]
[69]
Babcock, J.J.; Li, M. Deorphanizing the human transmembrane genome: A landscape of uncharacterized membrane proteins. Acta Pharmacol. Sin., 2014, 35(1), 11-23.
[http://dx.doi.org/10.1038/aps.2013.142] [PMID: 24241348]
[70]
Carpenter, E.P.; Beis, K.; Cameron, A.D.; Iwata, S. Overcoming the challenges of membrane protein crystallography. Curr. Opin. Struct. Biol., 2008, 18(5), 581-586.
[http://dx.doi.org/10.1016/j.sbi.2008.07.001] [PMID: 18674618]
[71]
Gopinath, T.; Weber, D.; Wang, S.; Larsen, E.; Veglia, G. Solid-state NMR of membrane proteins in lipid bilayers: To spin or not to spin? Acc. Chem. Res., 2021, 54(6), 1430-1439.
[http://dx.doi.org/10.1021/acs.accounts.0c00670] [PMID: 33655754]
[72]
Mitra, S.D.; Afonina, I.; Kline, K.A. Right place, right time: Focalization of membrane proteins in gram-positive bacteria. Trends Microbiol., 2016, 24(8), 611-621.
[http://dx.doi.org/10.1016/j.tim.2016.03.009] [PMID: 27117048]
[73]
Foot, N.; Henshall, T.; Kumar, S. Ubiquitination and the regulation of membrane proteins. Physiol. Rev., 2017, 97(1), 253-281.
[http://dx.doi.org/10.1152/physrev.00012.2016] [PMID: 27932395]
[74]
Marx, S.; Dal Maso, T.; Chen, J.W.; Bury, M.; Wouters, J.; Michiels, C.; Le Calvé, B. Transmembrane (TMEM) protein family members: Poorly characterized even if essential for the metastatic process. Semin. Cancer Biol., 2020, 60(1), 96-106.
[http://dx.doi.org/10.1016/j.semcancer.2019.08.018] [PMID: 31454669]
[75]
Wang, Y.; Zhang, S.; Li, F.; Zhou, Y.; Zhang, Y.; Wang, Z.; Zhang, R.; Zhu, J.; Ren, Y.; Tan, Y.; Qin, C.; Li, Y.; Li, X.; Chen, Y.; Zhu, F. Therapeutic target database 2020: Enriched resource for facilitating research and early development of targeted therapeutics. Nucleic Acids Res., 2020, 48(D1), D1031-D1041.
[PMID: 31691823]
[76]
Xue, W.; Fu, T.; Deng, S.; Yang, F.; Yang, J.; Zhu, F. Molecular mechanism for the allosteric inhibition of the human serotonin transporter by antidepressant escitalopram. ACS Chem. Neurosci., 2022, 13(3), 340-351.
[http://dx.doi.org/10.1021/acschemneuro.1c00694] [PMID: 35041375]
[77]
Alam, A.; Locher, K.P. Structure and mechanism of human ABC transporters. Annu. Rev. Biophys., 2023, 52(1), 275-300.
[http://dx.doi.org/10.1146/annurev-biophys-111622-091232] [PMID: 36737602]
[78]
Shvarev, D.; Januliene, D.; Moeller, A. Frozen motion: How cryo-EM changes the way we look at ABC transporters. Trends Biochem. Sci., 2022, 47(2), 136-148.
[http://dx.doi.org/10.1016/j.tibs.2021.11.008] [PMID: 34930672]
[79]
Hou, Z.; Gangjee, A.; Matherly, L.H. The evolving biology of the proton-coupled folate transporter: New insights into regulation, structure, and mechanism. FASEB J., 2022, 36(2), e22164.
[http://dx.doi.org/10.1096/fj.202101704R] [PMID: 35061292]
[80]
Kanai, Y. Amino acid transporter LAT1 (SLC7A5) as a molecular target for cancer diagnosis and therapeutics. Pharmacol. Ther., 2022, 230, 107964.
[http://dx.doi.org/10.1016/j.pharmthera.2021.107964] [PMID: 34390745]
[81]
Chiu, W.; Schmid, M.F.; Pintilie, G.D.; Lawson, C.L. Evolution of standardization and dissemination of cryo-EM structures and data jointly by the community, PDB, and EMDB. J. Biol. Chem., 2021, 296, 100560.
[http://dx.doi.org/10.1016/j.jbc.2021.100560] [PMID: 33744287]
[82]
Choy, B.C.; Cater, R.J.; Mancia, F.; Pryor, E.E.; Jr, A. 10-year meta-analysis of membrane protein structural biology: Detergents, membrane mimetics, and structure determination techniques. Biochim. Biophys. Acta Biomembr., 2021, 1863(3), 183533.
[http://dx.doi.org/10.1016/j.bbamem.2020.183533] [PMID: 33340490]
[83]
Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The protein data bank. Nucleic Acids Res., 2000, 28(1), 235-242.
[http://dx.doi.org/10.1093/nar/28.1.235] [PMID: 10592235]
[84]
Burley, S.K.; Bhikadiya, C.; Bi, C.; Bittrich, S.; Chen, L.; Crichlow, G.V.; Christie, C.H.; Dalenberg, K.; Di Costanzo, L.; Duarte, J.M.; Dutta, S.; Feng, Z.; Ganesan, S.; Goodsell, D.S.; Ghosh, S.; Green, R.K. Guranović V.; Guzenko, D.; Hudson, B.P.; Lawson, C.L.; Liang, Y.; Lowe, R.; Namkoong, H.; Peisach, E.; Persikova, I.; Randle, C.; Rose, A.; Rose, Y.; Sali, A.; Segura, J.; Sekharan, M.; Shao, C.; Tao, Y.P.; Voigt, M.; Westbrook, J.D.; Young, J.Y.; Zardecki, C.; Zhuravleva, M. RCSB Protein Data Bank: Powerful new tools for exploring 3D structures of biological macromolecules for basic and applied research and education in fundamental biology, biomedicine, biotechnology, bioengineering and energy sciences. Nucleic Acids Res., 2021, 49(D1), D437-D451.
[http://dx.doi.org/10.1093/nar/gkaa1038] [PMID: 33211854]
[85]
Gromiha, M.M.; Ou, Y.Y. Bioinformatics approaches for functional annotation of membrane proteins. Brief. Bioinform., 2014, 15(2), 155-168.
[http://dx.doi.org/10.1093/bib/bbt015] [PMID: 23524979]
[86]
del Alamo, D.; DeSousa, L.; Nair, R.M.; Rahman, S.; Meiler, J.; Mchaourab, H.S. Integrated AlphaFold2 and DEER investigation of the conformational dynamics of a pH-dependent APC antiporter. Proc. Natl. Acad. Sci. USA, 2022, 119(34), e2206129119.
[http://dx.doi.org/10.1073/pnas.2206129119] [PMID: 35969794]
[87]
Janaszkiewicz, A.; Tóth, Á.; Faucher, Q.; Martin, M.; Chantemargue, B.; Barin-Le Guellec, C.; Marquet, P.; Di Meo, F. Insights into the structure and function of the human organic anion transporter 1 in lipid bilayer membranes. Sci. Rep., 2022, 12(1), 7057.
[http://dx.doi.org/10.1038/s41598-022-10755-2] [PMID: 35488116]
[88]
Baek, M.; DiMaio, F.; Anishchenko, I.; Dauparas, J.; Ovchinnikov, S.; Lee, G.R.; Wang, J.; Cong, Q.; Kinch, L.N.; Schaeffer, R.D.; Millán, C.; Park, H.; Adams, C.; Glassman, C.R.; DeGiovanni, A.; Pereira, J.H.; Rodrigues, A.V.; van Dijk, A.A.; Ebrecht, A.C.; Opperman, D.J.; Sagmeister, T.; Buhlheller, C.; Pavkov-Keller, T.; Rathinaswamy, M.K.; Dalwadi, U.; Yip, C.K.; Burke, J.E.; Garcia, K.C.; Grishin, N.V.; Adams, P.D.; Read, R.J.; Baker, D. Accurate prediction of protein structures and interactions using a three-track neural network. Science, 2021, 373(6557), 871-876.
[http://dx.doi.org/10.1126/science.abj8754] [PMID: 34282049]
[89]
Doerr, S.; Majewski, M.; Pérez, A.; Krämer, A.; Clementi, C.; Noe, F.; Giorgino, T.; De Fabritiis, G.; Torch, M.D. A deep learning framework for molecular simulations. J. Chem. Theory Comput., 2021, 17(4), 2355-2363.
[http://dx.doi.org/10.1021/acs.jctc.0c01343] [PMID: 33729795]
[90]
Mulnaes, D.; Koenig, F.; Gohlke, H. TopSuite web server: A meta-suite for deep-learning-based protein structure and quality prediction. J. Chem. Inf. Model., 2021, 61(2), 548-553.
[http://dx.doi.org/10.1021/acs.jcim.0c01202] [PMID: 33464891]
[91]
Gao, M.; Zhou, H.; Skolnick, J. DESTINI: A deep-learning approach to contact-driven protein structure prediction. Sci. Rep., 2019, 9(1), 3514.
[http://dx.doi.org/10.1038/s41598-019-40314-1] [PMID: 30837676]
[92]
King, J.E.; Koes, D.R. SIDECHAINNET: An ALL‐ATOM protein structure dataset for machine learning. Proteins, 2021, 89(11), 1489-1496.
[http://dx.doi.org/10.1002/prot.26169] [PMID: 34213059]
[93]
Jensen, O.; Brockmöller, J.; Dücker, C. Identification of novel high-affinity substrates of OCT1 using machine learning-guided virtual screening and experimental validation. J. Med. Chem., 2021, 64(5), 2762-2776.
[http://dx.doi.org/10.1021/acs.jmedchem.0c02047] [PMID: 33606526]
[94]
Zhai, G.; Zhang, Z.; Dong, C. Mutagenesis and functional analysis of SotB: A multidrug transporter of the major facilitator superfamily from Escherichia coli. Front. Microbiol., 2022, 13, 1024639.
[http://dx.doi.org/10.3389/fmicb.2022.1024639] [PMID: 36386622]
[95]
Kabra, R.; Ingale, P.; Singh, S. Computationally designed synthetic peptides for transporter proteins imparts allostericity in Miltefosine resistant L. Major. Biochem. J., 2020, 477(10), 2007-2026.
[http://dx.doi.org/10.1042/BCJ20200176] [PMID: 32391551]
[96]
Xue, W.; Yang, F.; Wang, P.; Zheng, G.; Chen, Y.; Yao, X.; Zhu, F. What contributes to serotonin-norepinephrine reuptake inhibitors’ dual-targeting mechanism? the key role of transmembrane domain 6 in human serotonin and norepinephrine transporters revealed by molecular dynamics simulation. ACS Chem. Neurosci., 2018, 9(5), 1128-1140.
[http://dx.doi.org/10.1021/acschemneuro.7b00490] [PMID: 29300091]
[97]
Yin, J.; Sun, W.; Li, F.; Hong, J.; Li, X.; Zhou, Y.; Lu, Y.; Liu, M.; Zhang, X.; Chen, N.; Jin, X.; Xue, J.; Zeng, S.; Yu, L.; Zhu, F. VARIDT 1.0: Variability of drug transporter database. Nucleic Acids Res., 2020, 48(D1), D1042-D1050.
[http://dx.doi.org/10.1093/nar/gkz779] [PMID: 31495872]
[98]
Fu, T.; Li, F.; Zhang, Y.; Yin, J.; Qiu, W.; Li, X.; Liu, X.; Xin, W.; Wang, C.; Yu, L.; Gao, J.; Zheng, Q.; Zeng, S.; Zhu, F. VARIDT 2.0: Structural variability of drug transporter. Nucleic Acids Res., 2022, 50(D1), D1417-D1431.
[http://dx.doi.org/10.1093/nar/gkab1013] [PMID: 34747471]
[99]
Mak, L.; Marcus, D.; Howlett, A.; Yarova, G.; Duchateau, G.; Klaffke, W.; Bender, A.; Glen, R.C. Metrabase: A cheminformatics and bioinformatics database for small molecule transporter data analysis and (Q)SAR modeling. J. Cheminform., 2015, 7(1), 31.
[http://dx.doi.org/10.1186/s13321-015-0083-5] [PMID: 26106450]
[100]
Li, D.; Chen, L.; Li, Y.; Tian, S.; Sun, H.; Hou, T. ADMET evaluation in drug discovery. 13. Development of in silico prediction models for P-glycoprotein substrates. Mol. Pharm., 2014, 11(3), 716-726.
[http://dx.doi.org/10.1021/mp400450m] [PMID: 24499501]
[101]
Hazai, E.; Hazai, I.; Ragueneau-Majlessi, I.; Chung, S.P.; Bikadi, Z.; Mao, Q. Predicting substrates of the human breast cancer resistance protein using a support vector machine method. BMC Bioinformatics, 2013, 14(1), 130.
[http://dx.doi.org/10.1186/1471-2105-14-130] [PMID: 23586520]
[102]
Wang, X.; Zhu, X.; Ye, M.; Wang, Y.; Li, C.D.; Xiong, Y.; Wei, D.Q. STS-NLSP: A network-based label space partition method for predicting the specificity of membrane transporter substrates using a hybrid feature of structural and semantic similarity. Front. Bioeng. Biotechnol., 2019, 7, 306.
[http://dx.doi.org/10.3389/fbioe.2019.00306] [PMID: 31781551]
[103]
Alballa, M.; Aplop, F.; Butler, G. TranCEP: Predicting the substrate class of transmembrane transport proteins using compositional, evolutionary, and positional information. PLoS One, 2020, 15(1), e0227683.
[http://dx.doi.org/10.1371/journal.pone.0227683] [PMID: 31935244]
[104]
Nguyen, T.T.D.; Le, N.Q.K.; Ho, Q.T.; Phan, D.V.; Ou, Y.Y. Using word embedding technique to efficiently represent protein sequences for identifying substrate specificities of transporters. Anal. Biochem., 2019, 577(1), 73-81.
[http://dx.doi.org/10.1016/j.ab.2019.04.011] [PMID: 31022378]
[105]
Li, L.; Li, J.; Xiao, W.; Li, Y.; Qin, Y.; Zhou, S. Prediction the substrate specificities of membrane transport proteins based on support vector machine and hybrid features. IEEE/ACM Transact. Computat. Biol. Bioinformat., 2016, 13(5), 947-953.
[http://dx.doi.org/10.1109/TCBB.2015.2495140]
[106]
Mishra, N.K.; Chang, J.; Zhao, P.X. Prediction of membrane transport proteins and their substrate specificities using primary sequence information. PLoS One, 2014, 9(6), e100278.
[http://dx.doi.org/10.1371/journal.pone.0100278] [PMID: 24968309]
[107]
Rifaioglu, A.S.; Nalbat, E.; Atalay, V.; Martin, M.J.; Cetin-Atalay, R. Doğan, T. DEEPScreen: High performance drug–target interaction prediction with convolutional neural networks using 2-D structural compound representations. Chem. Sci. (Camb.), 2020, 11(9), 2531-2557.
[http://dx.doi.org/10.1039/C9SC03414E] [PMID: 33209251]
[108]
Lee, I.; Keum, J.; Nam, H. DeepConv-DTI: Prediction of drug-target interactions via deep learning with convolution on protein sequences. PLOS Comput. Biol., 2019, 15(6), e1007129.
[http://dx.doi.org/10.1371/journal.pcbi.1007129] [PMID: 31199797]
[109]
Hasan Mahmud, S.M.; Chen, W.; Jahan, H.; Dai, B.; Din, S.U.; Dzisoo, A.M. DeepACTION: A deep learning-based method for predicting novel drug-target interactions. Anal. Biochem., 2020, 610(1), 113978.
[http://dx.doi.org/10.1016/j.ab.2020.113978] [PMID: 33035462]
[110]
Kong, W.; Wang, W.; An, J. Prediction of 5-hydroxytryptamine transporter inhibitors based on machine learning. Comput. Biol. Chem., 2020, 87(1), 107303.
[http://dx.doi.org/10.1016/j.compbiolchem.2020.107303] [PMID: 32563857]
[111]
Kharangarh, S.; Sandhu, H.; Tangadpalliwar, S.; Garg, P. Predicting inhibitors for multidrug resistance associated protein-2 transporter by machine learning approach. Comb. Chem. High Throughput Screen., 2018, 21(8), 557-566.
[http://dx.doi.org/10.2174/1386207321666181024104822] [PMID: 30360705]
[112]
Lee, K.H.; Fant, A.D.; Guo, J.; Guan, A.; Jung, J.; Kudaibergenova, M.; Miranda, W.E.; Ku, T.; Cao, J.; Wacker, S.; Duff, H.J.; Newman, A.H.; Noskov, S.Y.; Shi, L. Toward reducing hERG affinities for DAT inhibitors with a combined machine learning and molecular modeling approach. J. Chem. Inf. Model., 2021, 61(9), 4266-4279.
[http://dx.doi.org/10.1021/acs.jcim.1c00856] [PMID: 34420294]
[113]
Khuri, N.; Deshmukh, S. Machine learning for classification of inhibitors of hepatic drug transporters. 17th IEEE Intl. Conf. Machine Learning Applicat, 2018, pp. 181-186.
[114]
Ibrahim, S.R.M.; Omar, A.M.; Muhammad, Y.A.; Alqarni, A.A.; Alshehri, A.M.; Mohamed, S.G.A.; Abdallah, H.M.; Elfaky, M.A.; Mohamed, G.A.; Xiao, J. Advances in fungal phenaloenones-natural metabolites with great promise: Biosynthesis, bioactivities, and an in silico evaluation of their potential as human glucose transporter 1 inhibitors. Molecules, 2022, 27(20), 6797.
[http://dx.doi.org/10.3390/molecules27206797] [PMID: 36296388]
[115]
Lane, T.R.; Urbina, F.; Zhang, X.; Fye, M.; Gerlach, J.; Wright, S.H.; Ekins, S. Machine learning models identify new inhibitors for human OATP1B1. Mol. Pharm., 2022, 19(11), 4320-4332.
[http://dx.doi.org/10.1021/acs.molpharmaceut.2c00662] [PMID: 36269563]
[116]
Iborra-Egea, O.; Santiago-Vacas, E.; Yurista, S.R.; Lupón, J.; Packer, M.; Heymans, S.; Zannad, F.; Butler, J.; Pascual-Figal, D.; Lax, A.; Núñez, J.; de Boer, R.A.; Bayés-Genís, A. Unraveling the molecular mechanism of action of empagliflozin in heart failure with reduced ejection fraction with or without diabetes. JACC Basic Transl. Sci., 2019, 4(7), 831-840.
[http://dx.doi.org/10.1016/j.jacbts.2019.07.010] [PMID: 31998851]
[117]
Krittanawong, C.; Johnson, K.W.; Rosenson, R.S.; Wang, Z.; Aydar, M.; Baber, U.; Min, J.K.; Tang, W.H.W.; Halperin, J.L.; Narayan, S.M. Deep learning for cardiovascular medicine: A practical primer. Eur. Heart J., 2019, 40(25), 2058-2073.
[http://dx.doi.org/10.1093/eurheartj/ehz056] [PMID: 30815669]
[118]
van der Hoek, S.A.; Borodina, I. Transporter engineering in microbial cell factories: The ins, the outs, and the in-betweens. Curr. Opin. Biotechnol., 2020, 66, 186-194.
[http://dx.doi.org/10.1016/j.copbio.2020.08.002] [PMID: 32927362]
[119]
Mutanda, I.; Sun, J.; Jiang, J.; Zhu, D. Bacterial membrane transporter systems for aromatic compounds: Regulation, engineering, and biotechnological applications. Biotechnol. Adv., 2022, 59, 107952.
[http://dx.doi.org/10.1016/j.biotechadv.2022.107952] [PMID: 35398204]
[120]
Nigam, S.K. The SLC22 transporter family: A paradigm for the impact of drug transporters on metabolic pathways, signaling, and disease. Annu. Rev. Pharmacol. Toxicol., 2018, 58(1), 663-687.
[http://dx.doi.org/10.1146/annurev-pharmtox-010617-052713] [PMID: 29309257]
[121]
Nigam, S.K.; Bush, K.T. Uraemic syndrome of chronic kidney disease: Altered remote sensing and signalling. Nat. Rev. Nephrol., 2019, 15(5), 301-316.
[http://dx.doi.org/10.1038/s41581-019-0111-1] [PMID: 30728454]
[122]
Bhatnagar, V.; Richard, E.L.; Wu, W.; Nievergelt, C.M.; Lipkowitz, M.S.; Jeff, J.; Maihofer, A.X.; Nigam, S.K. Analysis of ABCG2 and other urate transporters in uric acid homeostasis in chronic kidney disease: Potential role of remote sensing and signaling. Clin. Kidney J., 2016, 9(3), 444-453.
[http://dx.doi.org/10.1093/ckj/sfw010] [PMID: 27274832]
[123]
Nigam, S.K.; Granados, J.C. OAT, OATP, and MRP drug transporters and the remote sensing and signaling theory. Annu. Rev. Pharmacol. Toxicol., 2023, 63(1), 637-660.
[http://dx.doi.org/10.1146/annurev-pharmtox-030322-084058] [PMID: 36206988]
[124]
Jansen, J.; Jansen, K.; Neven, E.; Poesen, R.; Othman, A.; van Mil, A.; Sluijter, J.; Sastre Torano, J.; Zaal, E.A.; Berkers, C.R.; Esser, D.; Wichers, H.J.; van Ede, K.; van Duursen, M.; Burtey, S.; Verhaar, M.C.; Meijers, B.; Masereeuw, R. Remote sensing and signaling in kidney proximal tubules stimulates gut microbiome-derived organic anion secretion. Proc. Natl. Acad. Sci., 2019, 116(32), 16105-16110.
[http://dx.doi.org/10.1073/pnas.1821809116] [PMID: 31341083]
[125]
Thomik, T.; Wittig, I.; Choe, J.; Boles, E.; Oreb, M. An artificial transport metabolon facilitates improved substrate utilization in yeast. Nat. Chem. Biol., 2017, 13(11), 1158-1163.
[http://dx.doi.org/10.1038/nchembio.2457] [PMID: 28869594]
[126]
Li, W.; Ma, L.; Shen, X.; Wang, J.; Feng, Q.; Liu, L.; Zheng, G.; Yan, Y.; Sun, X.; Yuan, Q. Targeting metabolic driving and intermediate influx in lysine catabolism for high-level glutarate production. Nat. Commun., 2019, 10(1), 3337.
[http://dx.doi.org/10.1038/s41467-019-11289-4] [PMID: 31350399]
[127]
Kell, D.B.; Swainston, N.; Pir, P.; Oliver, S.G. Membrane transporter engineering in industrial biotechnology and whole cell biocatalysis. Trends Biotechnol., 2015, 33(4), 237-246.
[http://dx.doi.org/10.1016/j.tibtech.2015.02.001] [PMID: 25746161]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy