Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Differentiable biology: using deep learning for biophysics-based and data-driven modeling of molecular mechanisms

Abstract

Deep learning using neural networks relies on a class of machine-learnable models constructed using ‘differentiable programs’. These programs can combine mathematical equations specific to a particular domain of natural science with general-purpose, machine-learnable components trained on experimental data. Such programs are having a growing impact on molecular and cellular biology. In this Perspective, we describe an emerging ‘differentiable biology’ in which phenomena ranging from the small and specific (for example, one experimental assay) to the broad and complex (for example, protein folding) can be modeled effectively and efficiently, often by exploiting knowledge about basic natural phenomena to overcome the limitations of sparse, incomplete and noisy data. By distilling differentiable biology into a small set of conceptual primitives and illustrative vignettes, we show how it can help to address long-standing challenges in integrating multimodal data from diverse experiments across biological scales. This promises to benefit fields as diverse as biophysics and functional genomics.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Deep learning revolution.
Fig. 2: Neural network primitives.
Fig. 3: Differentiable programming fuses principles-based and data-driven modeling.
Fig. 4: Protein structure prediction vignette.
Fig. 5: PPI vignette.

Similar content being viewed by others

References

  1. Martín, A. et al. TensorFlow: large-scale machine learning on heterogeneous systems http://tensorflow.org/ (2015).

  2. Paszke, A. et al. Automatic differentiation in PyTorch. In 31st Conference on Neural Information Processing Systems (NIPS 2017) https://openreview.net/pdf?id=BJJsrmfCZ (2017).

  3. James, B., Roy, F., Peter, H., Matthew, B. & James, J. JAX: Autograd and XLA (Google, 2021).

  4. He, K., Zhang, X., Ren, S. & Sun, J. Deep residual learning for image recognition. Preprint at https://arxiv.org/abs/1512.03385 (2015).

  5. Russakovsky, O. et al. ImageNet Large Scale Visual Recognition Challenge. Int. J. Comput. Vis. 115, 211–252 (2015).

    Article  Google Scholar 

  6. Oei, R. W. et al. Convolutional neural network for cell classification using microscope images of intracellular actin networks. PLoS ONE 14, e0213626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Falk, T. et al. U-Net: deep learning for cell counting, detection, and morphometry. Nat. Methods 16, 67–70 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Serag, A. et al. Translational AI and deep learning in diagnostic pathology. Front. Med. 6, 185 (2019).

  9. Zhang, Z. et al. Pathologist-level interpretable whole-slide cancer diagnosis with deep learning. Nat. Mach. Intell. 1, 236–245 (2019).

    Article  Google Scholar 

  10. Vamathevan, J. et al. Applications of machine learning in drug discovery and development. Nat. Rev. Drug Discov. 18, 463–477 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cortes, C. & Vapnik, V. Support-vector networks. Machine Learn. 20, 273–297 (1995).

    Google Scholar 

  12. Tin Kam, H. Random decision forests. in Proceedings of the 3rd International Conference on Document Analysis and Recognition 278–282 (1995).

  13. Goodfellow, I., Bengio, Y. & Courville, A. Deep Learning (MIT Press, 2016).

  14. Zhang, R. Making convolutional networks shift-invariant again. Preprint at https://arxiv.org/abs/1904.11486 (2019).

  15. Rosenbaum, D. et al. Inferring a continuous distribution of atom coordinates from cryo-EM images using VAEs. Preprint at https://arxiv.org/abs/2106.14108 (2021).

  16. TensorFlow Core. Introducing the model garden for TensorFlow 2. TensorFlow Blog https://blog.tensorflow.org/2020/03/introducing-model-garden-for-tensorflow-2.html (2020).

  17. Wolf, T. et al. HuggingFace’s transformers: state-of-the-art natural language processing. Preprint at https://arxiv.org/abs/1910.03771 (2020).

  18. Ramsundar, B. et al. Deep Learning for the Life Sciences (O’Reilly Media, 2019).

  19. AlQuraishi, M. End-to-end differentiable learning of protein structure. Cell Syst. 8, 292–301 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gulshan, V. et al. Development and validation of a deep learning algorithm for detection of diabetic retinopathy in retinal fundus photographs. JAMA 316, 2402–2410 (2016).

    Article  PubMed  Google Scholar 

  21. Sadanandan, S. K., Ranefall, P., Guyader, S. L. & Wählby, C. Automated training of deep convolutional neural networks for cell segmentation. Sci. Rep. 7, 7860 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Gut, G., Herrmann, M. D. & Pelkmans, L. Multiplexed protein maps link subcellular organization to cellular states. Science 361, eaar7042 (2018).

    Article  PubMed  CAS  Google Scholar 

  23. Shorten, C. & Khoshgoftaar, T. M. A survey on image data augmentation for deep learning. J. Big Data 6, 60 (2019).

    Article  Google Scholar 

  24. Wang, S., Sun, S., Li, Z., Zhang, R. & Xu, J. Accurate de novo prediction of protein contact map by ultra-deep learning model. PLoS Comput. Biol. 13, e1005324 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Liu, Y., Palmedo, P., Ye, Q., Berger, B. & Peng, J. Enhancing evolutionary couplings with deep convolutional neural networks. Cell Syst. 6, 65–74 (2018).

    Article  PubMed  CAS  Google Scholar 

  26. Xu, J. Distance-based protein folding powered by deep learning. Proc. Natl Acad. Sci. USA 116, 16856–16865 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Senior, A. et al. Improved protein structure prediction using potentials from deep learning. Nature 577, 706–710 (2020).

  28. Torng, W. & Altman, R. B. High precision protein functional site detection using 3D convolutional neural networks. Bioinformatics 35, 1503–1512 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Gligorijevic, V. et al. Structure-based function prediction using graph convolutional networks. Preprint at bioRxiv https://doi.org/10.1101/786236 (2019).

  30. Wallach, I., Dzamba, M. & Heifets, A. AtomNet: a deep convolutional neural network for bioactivity prediction in structure-based drug discovery. Preprint at https://arxiv.org/abs/1510.02855 (2015).

  31. Gomes, J., Ramsundar, B., Feinberg, E. N. & Pande, V. S. Atomic convolutional networks for predicting protein–ligand binding affinity. Preprint at https://arxiv.org/abs/1703.10603 (2017).

  32. Benos, P. V., Lapedes, A. S. & Stormo, G. D. Is there a code for protein–DNA recognition? Probab(ilistical)ly…. BioEssays 24, 466–475 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Alipanahi, B., Delong, A., Weirauch, M. T. & Frey, B. J. Predicting the sequence specificities of DNA- and RNA-binding proteins by deep learning. Nat. Biotechnol. 33, 831–838 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Avsec, Z. et al. Deep learning at base-resolution reveals motif syntax of the cis-regulatory code. Preprint at bioRxiv https://doi.org/10.1101/737981 (2019).

  35. Wu, Z. et al. A comprehensive survey on graph neural networks. Preprint at https://arxiv.org/abs/1901.00596 (2019).

  36. Segler, M. H. S., Preuss, M. & Waller, M. P. Planning chemical syntheses with deep neural networks and symbolic AI. Nature 555, 604–610 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Bouatta, N., Sorger, P. & AlQuraishi, M. Protein structure prediction by AlphaFold2: are attention and symmetries all you need? Acta Crystallogr. D Struct. Biol. 77, 982–991 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature https://doi.org/10.1038/s41586-021-03819-2 (2021).

  39. Muzio, G., O’Bray, L. & Borgwardt, K. Biological network analysis with deep learning. Brief. Bioinform. 22, 1515–1530 (2021).

    Article  PubMed  Google Scholar 

  40. Chowdhury, R. et al. Single-sequence protein structure prediction using language models from deep learning. Preprint at bioRxiv https://doi.org/10.1101/2021.08.02.454840 (2021).

  41. Hall, B. Lie Groups, Lie Algebras, and Representations: An Elementary Introduction (Springer, 2004).

  42. Cohen, T. S., Geiger, M. & Weiler, M. A general theory of equivariant CNNs on homogeneous spaces. In Advances in Neural Information Processing Systems vol. 32 (Curran, 2019).

  43. Weiler, M., Geiger, M., Welling, M., Boomsma, W. & Cohen, T. S. 3D Steerable CNNs: learning rotationally equivariant features in volumetric data. In Advances in Neural Information Processing Systems vol. 31 (Curran, 2018).

  44. Gao, M. & Skolnick, J. Structural space of protein–protein interfaces is degenerate, close to complete, and highly connected. Proc. Natl Acad. Sci. USA 107, 22517–22522 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gainza, P. et al. Deciphering interaction fingerprints from protein molecular surfaces using geometric deep learning. Nat. Methods 17, 184–192 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Akbar, R. et al. A compact vocabulary of paratope-epitope interactions enables predictability of antibody-antigen binding. Cell Rep. 34, 108856 (2021).

    Article  CAS  PubMed  Google Scholar 

  47. Cunningham, J., Koytiger, G., Sorger, P. K. & AlQuraishi, M. Biophysical prediction of protein–peptide interactions and signaling networks using machine learning. Nat. Methods 17, 175–183 (2020).

  48. Townshend, R., Bedi, R., Suriana, P. & Dror, R. End-to-end learning on 3D protein structure for interface prediction. In Advances in Neural Information Processing Systems vol. 32 (Curran, 2019).

  49. Paggi, J. M. et al. Leveraging non-structural data to predict structures of protein–ligand complexes. Preprint at bioRxiv https://doi.org/10.1101/2020.06.01.128181 (2020).

  50. Berg, S. et al. ilastik: interactive machine learning for (bio)image analysis. Nat. Methods https://doi.org/10.1038/s41592-019-0582-9 (2019).

  51. Krueger, R. et al. Facetto: combining unsupervised and supervised learning for hierarchical phenotype analysis in multi-channel image data. IEEE Trans. Vis. Comput. Graph. https://doi.org/10.1109/TVCG.2019.2934547 (2019).

  52. Bialek, W. Biophysics: Searching for Principles (Princeton Univ. Press, 2012).

  53. Nguyen, T. H. et al. Bayesian analysis of isothermal titration calorimetry for binding thermodynamics. PLoS ONE 13, e0203224 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Chen, T. Q., Rubanova, Y., Bettencourt, J. & Duvenaud, D. K. Neural ordinary differential equations. In Advances in Neural Information Processing Systems (eds. Bengio, S. et al.) 6571–6583 (Curran, 2018).

  55. Yuan, B. et al. CellBox: interpretable machine learning for perturbation biology with application to the design of cancer combination therapy. Cell Syst. 12, 128–140 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Baek, M. et al. Accurate prediction of protein structures and interactions using a three-track neural network. Science https://doi.org/10.1126/science.abj8754 (2021).

  57. Branden, C. & Tooze, J. Introduction to Protein Structure (Garland Science, 1999).

  58. Parsons, J., Holmes, J. B., Rojas, J. M., Tsai, J. & Strauss, C. E. M. Practical conversion from torsion space to Cartesian space for in silico protein synthesis. J. Comput. Chem. 26, 1063–1068 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. AlQuraishi, M. ProteinNet: a standardized data set for machine learning of protein structure. BMC Bioinformatics 20, 311 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Fuchs, F., Worrall, D., Fischer, V. & Welling, M. SE(3)-transformers: 3D roto-translation equivariant attention networks. In Advances in Neural Information Processing Systems vol. 33 1970–1981 (Curran, 2020).

  61. Devlin, J., Chang, M.-W., Lee, K. & Toutanova, K. BERT: pre-training of deep bidirectional transformers for language understanding. In Proc. 2019 Conference of the North American Chapter of the Association for Computational Linguistics: Human Language Technologies, NAACL-HLT 2019, Minneapolis, MN, USA, June 2-7, 2019, Volume 1 (Long and Short Papers) (eds. Burstein, J., Doran, C. & Solorio, T.) 4171–4186 (Association for Computational Linguistics, 2019).

  62. Vaswani, A. et al. Attention is all you need. In Advances in Neural Information Processing Systems vol. 30 (Curran, 2017).

  63. Lee, H.-J. & Zheng, J. J. PDZ domains and their binding partners: structure, specificity, and modification. Cell Commun. Signal. 8, 8 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Song, J., Hao, Y., Du, Z., Wang, Z. & Ewing, R. M. Identifying novel protein complexes in cancer cells using epitope-tagging of endogenous human genes and affinity-purification mass spectrometry. J. Proteome Res. 11, 5630–5641 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chatr-aryamontri, A. et al. The BioGRID interaction database: 2017 update. Nucleic Acids Res. 45, D369–D379 (2017).

    Article  CAS  PubMed  Google Scholar 

  66. Luck, K. et al. A reference map of the human binary protein interactome. Nature 580, 402–408 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  68. Martins, A. & Astudillo, R. From Softmax to Sparsemax: a sparse model of attention and multi-label classification. In International Conference on Machine Learning 1614–1623 (PMLR, 2016).

  69. Rasmussen, C. E. & Williams, C. K. I. Gaussian Processes for Machine Learning (MIT Press, 2005).

  70. Maclaurin, D., Duvenaud, D. & Adams, R. Gradient-based hyperparameter optimization through reversible learning. In International Conference on Machine Learning 2113–2122 (PMLR, 2015).

  71. Lorraine, J. & Duvenaud, D. Stochastic hyperparameter optimization through hypernetworks. Preprint at https://arxiv.org/abs/1802.09419 (2018).

  72. Burgess, D. J. Spatial transcriptomics coming of age. Nat. Rev. Genet. 20, 317 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Reddy, R. J. et al. Early signaling dynamics of the epidermal growth factor receptor. Proc. Natl Acad. Sci. USA 113, 3114–3119 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Maier, T., Güell, M. & Serrano, L. Correlation of mRNA and protein in complex biological samples. FEBS Lett. 583, 3966–3973 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Garnett, M. J. et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Costello, J. C. et al. A community effort to assess and improve drug sensitivity prediction algorithms. Nat. Biotechnol. 32, 1202–1212 (2014).

  78. Aldridge, B. B., Burke, J. M., Lauffenburger, D. A. & Sorger, P. K. Physicochemical modelling of cell signalling pathways. Nat. Cell Biol. 8, 1195–1203 (2006).

    Article  CAS  PubMed  Google Scholar 

  79. Rackauckas, C. et al. Universal differential equations for scientific machine learning. Preprint at https://arxiv.org/abs/2001.04385 (2020).

  80. Yang, J., Li, A., Li, Y., Guo, X. & Wang, M. A novel approach for drug response prediction in cancer cell lines via network representation learning. Bioinformatics 35, 1527–1535 (2019).

    Article  CAS  PubMed  Google Scholar 

  81. Neil, D., Pfeiffer, M. & Liu, S.-C. Phased LSTM: accelerating recurrent network training for long or event-based sequences. In Advances in Neural Information Processing Systems vol. 29 (Curran, 2016).

  82. Eydgahi, H. et al. Properties of cell death models calibrated and compared using Bayesian approaches. Mol. Syst. Biol. 9, 644 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Dillon, J. V. et al. TensorFlow distributions. Preprint at https://arxiv.org/abs/1711.10604 (2017).

  84. Bingham, E. et al. Pyro: deep universal probabilistic programming. J. Mach. Learn. Res. 20, 1–6 (2019).

    Google Scholar 

  85. Hafner, M., Niepel, M. & Sorger, P. K. Alternative drug sensitivity metrics improve preclinical cancer pharmacogenomics. Nat. Biotechnol. 35, 500–502 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Saar-Tsechansky, M. & Provost, F. Handling missing values when applying classification models. J. Mach. Learn. Res. 8, 1623–1657 (2007).

    Google Scholar 

  87. Bepler, T. & Berger, B. Learning the protein language: evolution, structure, and function. Cell Syst. 12, 654–669 (2021).

    Article  CAS  PubMed  Google Scholar 

  88. Bepler, T. & Berger, B. Learning protein sequence embeddings using information from structure. In International Conference on Learning Representations (2019).

  89. Alley, E. C., Khimulya, G., Biswas, S., AlQuraishi, M. & Church, G. M. Unified rational protein engineering with sequence-based deep representation learning. Nat. Methods 16, 1315–1322 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Elnaggar, A. et al. ProtTrans: towards cracking the language of lifes code through self-supervised deep learning and high performance computing. IEEE Trans. Pattern Anal. Mach. Intel. https://doi.org/10.1109/TPAMI.2021.3095381 (2021).

  91. Madani, A. et al. ProGen: language modeling for protein generation. Preprint at https://arxiv.org/abs/2004.03497 (2020).

  92. Rives, A. et al. Biological structure and function emerge from scaling unsupervised learning to 250 million protein sequences. Proc. Natl Acad. Sci. USA 118, e2016239118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Biswas, S., Khimulya, G., Alley, E. C., Esvelt, K. M. & Church, G. M. Low-N protein engineering with data-efficient deep learning. Nat. Methods 18, 389–396 (2021).

    Article  CAS  PubMed  Google Scholar 

  94. Weißenow, K., Heinzinger, M. & Rost, B. Protein language model embeddings for fast, accurate, alignment-free protein structure prediction. Preprint at bioRxiv https://doi.org/10.1101/2021.07.31.454572 (2021).

  95. Bileschi, M. L. et al. Using deep learning to annotate the protein universe. Preprint at bioRxiv https://doi.org/10.1101/626507 (2019).

  96. Lai, B. & Xu, J. Accurate protein function prediction via graph attention networks with predicted structure information. Preprint at bioRxiv https://doi.org/10.1101/2021.06.16.448727 (2021).

  97. Gligorijević, V. et al. Structure-based protein function prediction using graph convolutional networks. Nat. Commun. 12, 3168 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Rao, R. et al. MSA Transformer. Preprint at bioRxiv https://doi.org/10.1101/2021.02.12.430858 (2021).

  99. Sterling, T. & Irwin, J. J. ZINC 15—ligand discovery for everyone. J. Chem. Inf. Model. 55, 2324–2337 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hu, W. et al. Strategies for pre-training graph neural networks. In International Conference on Learning Representations (2019).

  101. Liu, S., Demirel, M. F. & Liang, Y. N-gram graph: simple unsupervised representation for graphs, with applications to molecules. In Advances in Neural Information Processing Systems vol. 32 (Curran, 2019).

  102. Chithrananda, S., Grand, G. & Ramsundar, B. ChemBERTa: large-scale self-supervised pretraining for molecular property prediction. Preprint at https://arxiv.org/abs/2010.09885 (2020).

  103. Wang, Y., Wang, J., Cao, Z. & Farimani, A. B. MolCLR: molecular contrastive learning of representations via graph neural networks. Preprint at https://arxiv.org/abs/2102.10056 (2021).

  104. Zhu, J. et al. Dual-view molecule pre-training. Preprint at https://arxiv.org/abs/2106.10234 (2021).

  105. Goodfellow, I. et al. Generative adversarial nets. In Advances in Neural Information Processing Systems (eds. Ghahramani, Z. et al.) 2672–2680 (Curran, 2014).

  106. Kingma, D. P. & Welling, M. Auto-encoding variational Bayes. Preprint at https://arxiv.org/abs/1312.6114 (2013).

  107. Kobyzev, I., Prince, S. J. D. & Brubaker, M. A. Normalizing flows: an introduction and review of current methods. IEEE Trans. Pattern Anal. Mach. Intell. https://doi.org/10.1109/TPAMI.2020.2992934 (2020).

  108. Sohl-Dickstein, J., Weiss, E. A., Maheswaranathan, N. & Ganguli, S. Deep unsupervised learning using nonequilibrium thermodynamics. Preprint at https://arxiv.org/abs/1503.03585 (2015).

  109. Karras, T., Aila, T., Laine, S. & Lehtinen, J. Progressive growing of GANs for improved quality, stability, and variation. In International Conference on Learning Representations (2018).

  110. De Cao, N. & Kipf, T. MolGAN: an implicit generative model for small molecular graphs. Preprint at https://arxiv.org/abs/1805.11973 (2018).

  111. Gómez-Bombarelli, R. et al. Automatic chemical design using a data-driven continuous representation of molecules. ACS Cent. Sci. 4, 268–276 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Zhavoronkov, A. et al. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat. Biotechnol. 37, 1038–1040 (2019).

    Article  CAS  PubMed  Google Scholar 

  113. Killoran, N., Lee, L. J., Delong, A., Duvenaud, D. & Frey, B. J. Generating and designing DNA with deep generative models. Preprint at https://arxiv.org/abs/1712.06148 (2017).

  114. Anand, N., Eguchi, R. & Huang, P.-S. Fully differentiable full-atom protein backbone generation. In International Conference on Learning Representations Workshop (2019).

  115. Ingraham, J., Garg, V., Barzilay, R. & Jaakkola, T. Generative models for graph-based protein design. In Advances in Neural Information Processing Systems vol. 32 (Curran, 2019).

  116. Marouf, M. et al. Realistic in silico generation and augmentation of single-cell RNA-seq data using generative adversarial networks. Nat. Commun. 11, 166 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Johnson-Roberson, M. et al. Driving in the matrix: can virtual worlds replace human-generated annotations for real world tasks? Preprint at https://arxiv.org/abs/1610.01983 (2017).

  118. Martin, R. M. Electronic Structure: Basic Theory and Practical Methods (Cambridge University Press, 2008).

  119. Smith, J. S., Isayev, O. & Roitberg, A. E. ANI-1: an extensible neural network potential with DFT accuracy at force field computational cost. Chem. Sci. 8, 3192–3203 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Brockherde, F. et al. Bypassing the Kohn–Sham equations with machine learning. Nat. Commun. 8, 872 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Zhang, L., Han, J., Wang, H., Car, R., & Weinan, E. Deep potential molecular dynamics: a scalable model with the accuracy of quantum mechanics. Phys. Rev. Lett. 120, 143001 (2018).

    Article  CAS  PubMed  Google Scholar 

  122. OpenAI et al. Solving Rubik’s cube with a robot hand. Preprint at https://arxiv.org/abs/1910.07113 (2019).

  123. Kulkarni, T. D., Whitney, W. F., Kohli, P. & Tenenbaum, J. B. Deep convolutional inverse graphics network. in Proc. 28th International Conference on Neural Information Processing Systems Vol. 2, 2539–2547 (MIT Press, 2015).

  124. Carreira-Perpinan, M. A. & Hinton, G. E. On contrastive divergence learning. Aistats 10, 33–40 (2005).

    Google Scholar 

  125. Jumper, J. M., Faruk, N. F., Freed, K. F. & Sosnick, T. R. Trajectory-based training enables protein simulations with accurate folding and Boltzmann ensembles in CPU-hours. PLoS Comput. Biol. 14, e1006578 (2018).

  126. Ingraham, J., Riesselman, A., Sander, C. & Marks, D. Learning protein structure with a differentiable simulator. In International Conference on Learning Representations (2019).

  127. Wu, J. et al. EBM-Fold: fully-differentiable protein folding powered by energy-based models. Preprint at https://arxiv.org/abs/2105.04771 (2021).

  128. Walker, S. G. in Bayesian Nonparametrics (eds. Holmes, C., Hjort, N. L., Müller, P. & Walker, S. G.) 22–34 (Cambridge Univ. Press, 2010).

  129. Rezende, D. J., Mohamed, S. & Wierstra, D. Stochastic backpropagation and approximate inference in deep generative models. in Proceedings of the 31st International Conference on Machine Learning Vol. 32, II-1278–II-1286 (JMLR.org, 2014).

  130. Sutton, R. S. & Barto, A. G. Reinforcement Learning: An Introduction (MIT Press, 2018).

  131. Suarez, J., Du, Y., Mordach, I. & Isola, P. Neural MMO v1.3: a massively multiagent game environment for training and evaluating neural networks. In Proc. 19th International Conference on Autonomous Agents and MultiAgent Systems 2020–2022 (International Foundation for Autonomous Agents and Multiagent Systems, 2020).

  132. Vinyals, O. et al. Grandmaster level in StarCraft II using multi-agent reinforcement learning. Nature 575, 350–354 (2019).

    Article  CAS  PubMed  Google Scholar 

  133. Mikulak-Klucznik, B. et al. Computational planning of the synthesis of complex natural products. Nature https://doi.org/10.1038/s41586-020-2855-y (2020).

  134. Eastman, P., Shi, J., Ramsundar, B. & Pande, V. S. Solving the RNA design problem with reinforcement learning. PLoS Comput. Biol. 14, e1006176 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Webb, S. Deep learning for biology. Nature 554, 555–557 (2018).

    Article  CAS  PubMed  Google Scholar 

  136. Cho, J., Lee, K., Shin, E., Choy, G. & Do, S. How much data is needed to train a medical image deep learning system to achieve necessary high accuracy? Preprint at https://arxiv.org/abs/1511.06348 (2016).

  137. Zhou, J. et al. Graph neural networks: a review of methods and applications. Preprint at https://arxiv.org/abs/1812.08434 (2021).

  138. Wu, Z. et al. A comprehensive survey on graph neural networks. IEEE Trans. Neural Netw. Learn. Syst. 32, 4–24 (2021).

    Article  PubMed  Google Scholar 

  139. Bowman, S. R. et al. Generating sentences from a continuous space. In Proc. 20th SIGNLL Conference on Computational Natural Language Learning 10–21 (Association for Computational Linguistics, 2016).

  140. Lample, G. & Charton, F. Deep learning for symbolic mathematics. In International Conference for Learning Representations (2020).

  141. Grefenstette, E., Hermann, K. M., Suleyman, M. & Blunsom, P. Learning to transduce with unbounded memory. In Advances in Neural Information Processing Systems vol. 28 (Curran, 2015).

  142. Grover, A., Wang, E., Zweig, A. & Ermon, S. Stochastic optimization of sorting networks via continuous relaxations. In International Conference on Learning Representations (2018).

  143. Graves, A. Adaptive computation time for recurrent neural networks. Preprint at https://arxiv.org/abs/1603.08983 (2016).

  144. Trask, A. et al. Neural arithmetic logic units. In Advances in Neural Information Processing Systems vol. 31 (Curran, 2018).

  145. Jin, W., Barzilay, R. & Jaakkola, T. Junction tree variational autoencoder for molecular graph generation. In International Conference on Machine Learning 2323–2332 (PMLR, 2018).

  146. Amodei, D. & Hernandez, D. AI and compute. Heruntergeladen Von Httpsblog Openai Comaiand-Compute (2018).

  147. Weld, D. S. & Bansal, G. The challenge of crafting intelligible intelligence. Commun. ACM 62, 70–79 (2019).

    Article  Google Scholar 

  148. Chakraborty, S. et al. Interpretability of deep learning models: a survey of results. in 2017 IEEE SmartWorld, Ubiquitous Intelligence Computing, Advanced Trusted Computed, Scalable Computing Communications, Cloud Big Data Computing, Internet of People and Smart City Innovation (SmartWorld/SCALCOM/UIC/ATC/CBDCom/IOP/SCI) 1–6 (IEEE, 2017).

  149. Godfrey, J. J., Holliman, E. C. & McDaniel, J. SWITCHBOARD: telephone speech corpus for research and development. In Proc. 1992 IEEE international Conference on Acoustics, Speech and Signal Processing Vol. 1, 517–520 (IEEE Computer Society, 1992).

  150. Han, K. J., Chandrashekaran, A., Kim, J. & Lane, I. The CAPIO 2017 conversational speech recognition system. Preprint at https://arxiv.org/abs/1801.00059 (2018).

  151. Schütt, K. T. et al. (eds.) Machine Learning Meets Quantum Physics (Springer, 2020).

  152. Kryshtafovych, A., Schwede, T., Topf, M., Fidelis, K. & Moult, J. Critical assessment of methods of protein structure prediction (CASP)—Round XIII. Proteins 87, 1011–1020 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank N. Bouatta for comments on early versions of this manuscript. This work is supported by DARPA PANACEA program grant HR00111920022 and NCI/NIH grant U54-CA225088 to P.K.S.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Mohammed AlQuraishi or Peter K. Sorger.

Ethics declarations

Competing interests

P.K.S. is a member of the scientific advisory board or board of directors of Glencoe Software, Applied Biomath, RareCyte and NanoString and a consultant to Montai Health and Merck; he has equity in several of these companies. P.K.S. declares that none of these relationships are directly or indirectly related to the content of this manuscript. M.A.Q. is a member of the scientific advisory board of FL2021-002, a Foresite Labs company, and consults for Interline Therapeutics.

Additional information

Peer review information Arunima Singh was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

AlQuraishi, M., Sorger, P.K. Differentiable biology: using deep learning for biophysics-based and data-driven modeling of molecular mechanisms. Nat Methods 18, 1169–1180 (2021). https://doi.org/10.1038/s41592-021-01283-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01283-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing