Skip to content

frederikkemarin/BEND

Repository files navigation

🧬 BEND - Benchmarking DNA Language Models on Biologically Meaningful Tasks

Stars License Documentation Status

The BEND preprint is available here:

"BEND: BENCHMARKING DNA LANGUAGE MODELS ON BIOLOGICALLY MEANINGFUL TASKS"

Frederikke Isa Marin, Felix Teufel, Marc Horlacher, Dennis Madsen, Dennis Pultz, Ole Winther, Wouter Boomsma

Documentation

Documentation for the BEND code repository.

Data

All data is available for download here

The data can be downloaded via a script, see section 2.5

Tutorial

1. Data format

The data for each task is stored as a bed file. This file includes the genomic coordinates for each sample, as well as its split membership and potentially a label. Together with a reference genome, the file is used to extract the DNA sequences for training. Labels that are too complex to be stored in a column in the text-based bed file are stored in a hdf5 file. The two files share their index, so that sample i in the bed file matches record i in the hdf5 file.

bed is a tab-separated format that can be read like a regular table. All our task files include a column split, and optionally label. If label is missing, the labels are found in the hdf5 file of the same name.

chromosome	start	end     split	label
chr1	    1055037	1055849	train	1
chr3	    1070026	1070436	valid	0

2. Setup

We recommend installing BEND in a conda environment with Python 3.10.

  1. Clone the BEND repository: git clone https://github.com/frederikkemarin/BEND.git
  2. Change to the BEND directory: cd BEND
  3. Install the requirements: pip install -r requirements.txt
  4. Install BEND in development mode: pip install -e .
  5. Download the data: python scripts/download_bend.py

3. Computing embeddings

For training downstream models, it is practical to precompute and save the embeddings to avoid recomputing them at each epoch. As embeddings can grow large when working with genomes, we use Webdataset tar.gz files as the format. Firstly download the desired data from the data folder and place it in BEND/ (for ease of use maintain the same folder structure). To precompute the embeddings for all models and tasks, run :

python scripts/precompute_embeddings.py 

This script automatically calls the hydra config file at /../conf/embedding/embed.yaml.

By default all embeddings are generated for all tasks. To alter the tasks/model for which to compute the embeddings, please alter the tasks and/or the models list in the config file (under ```hydra.sweeper``) or override the behaviour from the commandline in the following manner:

python scripts/precompute_embeddings.py model=resnetlm,awdlstm task=gene_finding,enhancer_annotation

Train, validation and test embeddings are saved in chunks of (default) 50,000. To parallelize embeddings generation, you can call precompute_embeddings.py as above multiple times, but add additional arguments of the form chunk=[10,11,12] splits=[train,valid] to the individual calls in order to only compute specific chunks in a given call. If these arguments are not provided, the command will default to computing all chunks and splits.

Embedders overview

If you need to make embeddings for other purposes than preparing downstream task data, bend.embedders contains wrapper classes around the individual models. Each embedder takes a path (or name, if available on HuggingFace) of a checkpoint as the first argument, and provides an embed() method that takes a list of sequences and returns a list of embeddings.
Embedders have a default-true argument remove_special_tokens=True in embed() that removes any [CLS], [SEP] tokens from the returned embeddings. For models that return less embedding vectors than their number of input nucleotides, embeddings can be upsampled to the original input sequence length using the upsample_embeddings=True argument in embed().

Embedder Reference Models Info
DNABertEmbedder Ji et al. 4 different k-mer tokenizations available has an additional argument kmer=6 to specify the k-mer size.
NucleotideTransformerEmbedder Dalla-Torre et al. 8 different models available
ConvNetEmbedder BEND 1 model available A baseline LM used in BEND.
AWDLSTMEmbedder BEND 1 model available A baseline LM used in BEND.
GPNEmbedder Benegas et al. Models trained on A. thaliana and Brassicales available This LM was not evaluated in BEND as it was not trained on the human genome.
GENALMEmbedder Fishman et al. 8 different models available
HyenaDNAEmbedder Nguyen et al. 5 different models available Experimental integration. Requires Git LFS to be installed to automatically download checkpoints. Instead of the HF checkpoint name, the argument when instantiating needs to be of the format path/to/save/checkpoints/checkpoint_name
DNABert2Embedder Zhou et al. 1 model available
GROVEREmbedder Sanabria et al. 1 model available The original BPE tokenizer is not available, so we apply MaxMatch for segmentation of the input sequence into tokens.

All embedders can be used as follows:

from bend.embedders import NucleotideTransformerEmbedder

# load the embedder with a valid checkpoint name or path
embedder = NucleotideTransformerEmbedder('InstaDeepAI/nucleotide-transformer-2.5b-multi-species')

# embed a list of sequences
embeddings = embedder.embed(['AGGATGCCGAGAGTATATGGGA', 'CCCAACCGAGAGTATATGTTAT'])
# or just call directly to embed a single sequence
embedding = embedder('AGGATGCCGAGAGTATATGGGA') 

# This requires git LFS and will automatically download the checkpoint, if not already present
from bend.embedders import HyenaDNAEmbedder
embedder = HyenaDNAEmbedder('pretrained_models/hyenadna/hyenadna-tiny-1k-seqlen')

4. Evaluating models

Training and evaluating supervised models

It is first required that the above step (computing the embeddings) is completed. The embeddings should afterwards be located in BEND/data/{task_name}/{embedder}/*tar.gz

To run a downstream task run (from BEND/):

python scripts/train_on_task.py --config-name {tasl}

By default the task is run on all embeddings. To alter this either modify the config file or change the settings from the commandline E.g. to run gene finding on all embeddings the commandline is:

python scripts/train_on_task.py --config-name gene_finding

To run only on resnetlm and awdlstm embeddings:

python scripts/train_on_task.py --config-name gene_finding embedder=resnetlm,awdlstm

The full list of current task names are :

  • gene_finding
  • enhancer_annotation
  • variant_effects
  • histone_modification
  • chromatin_accessibility
  • cpg_methylation

And the list of available embedders/models used for training on the tasks are :

  • awdlstm
  • resnetlm
  • nt_transformer_ms
  • nt_transformer_human_ref
  • dnabert6
  • resnet_supervised
  • onehot
  • nt_transformer_1000g
  • dnabert2
  • gena-lm-bigbird-base-t2t
  • gena-lm-bert-large-t2
  • hyenadna-large-1m
  • hyenadna-tiny-1k
  • hyenadna-small-32k
  • hyenadna-medium-160k
  • grover

The train_on_task.py script calls a trainer class bend.utils.task_trainer. All configurations required to adapt these 2 scripts to train on a specific task (input data, downstream model, parameters, evaluation metric etc.) are specified in the task specific hydra config files stored in the conf directory. This minimizes the changes required to the scripts in order to introduce a potential new task.

The results of a run can be found at :

BEND/downstream_tasks/{task_name}/{embedder}/

If desired, the config files can be modified to change parameters, output/input directory etc.

Unsupervised tasks

For unsupervised prediction of variant effects, embeddings don't have to be precomputed and stored. Embeddings are generated and directly evaluated using

python3 scripts/predict_variant_effects.py {variant_file_name}.bed {output_file_name}.csv {model_type} {path_to_checkpoint} {path_to_reference_genome_fasta} --embedding_idx {position_of_embedding}

There are two variant effect prediction tasks available for {variant_file_name}: Variants with expression effect (eQTLs) in variant_effects_expression.bed and disease-causing variants in variant_effects_disease.bed.

A notebook with an example of how to run the script and evaluate the results can be found in examples/unsupervised_variant_effects.ipynb. To run all models, you can use the script scripts/run_variant_effects.sh.


Extending BEND

Adding a new embedder

All embedders are defined in bend/utils/embedders.py and inherit BaseEmbedder. A new embedder needs to implement load_model, which should set up all required attributes of the class and handle loading the model checkpoint into memory. It also needs to implement embed, which takes a list of sequences, and returns a list of embedding matrices formatted as numpy arrays. The embed method should be able to handle sequences of different lengths.

Adding a new task

As the first step, the data for a new task needs to be formatted in the bed-based format. If necessary, a split and label column should be included. The next step is to add new config files to ../conf/supervised_tasks. You should create a new directory named after the task, and add a config file for each embedder you want to evaluate. The config files should be named after the embedder.


Citation Guidelines

The datasets included in BEND were collected from a variety of sources. When you use any of the datasets, please ensure to correctly cite the respective original publications describing each dataset.

Gene finding (GENCODE)

@article{frankish_gencode_2021,
title = {{GENCODE} 2021},
volume = {49},
issn = {0305-1048},
url = {https://doi.org/10.1093/nar/gkaa1087},
doi = {10.1093/nar/gkaa1087},
number = {D1},
urldate = {2022-09-26},
journal = {Nucleic Acids Research},
author = {Frankish, Adam and Diekhans, Mark and Jungreis, Irwin and Lagarde, Julien and Loveland, Jane E and Mudge, Jonathan M and Sisu, Cristina and Wright, James C and Armstrong, Joel and Barnes, If and Berry, Andrew and Bignell, Alexandra and Boix, Carles and Carbonell Sala, Silvia and Cunningham, Fiona and Di Domenico, Tomás and Donaldson, Sarah and Fiddes, Ian T and García Girón, Carlos and Gonzalez, Jose Manuel and Grego, Tiago and Hardy, Matthew and Hourlier, Thibaut and Howe, Kevin L and Hunt, Toby and Izuogu, Osagie G and Johnson, Rory and Martin, Fergal J and Martínez, Laura and Mohanan, Shamika and Muir, Paul and Navarro, Fabio C P and Parker, Anne and Pei, Baikang and Pozo, Fernando and Riera, Ferriol Calvet and Ruffier, Magali and Schmitt, Bianca M and Stapleton, Eloise and Suner, Marie-Marthe and Sycheva, Irina and Uszczynska-Ratajczak, Barbara and Wolf, Maxim Y and Xu, Jinuri and Yang, Yucheng T and Yates, Andrew and Zerbino, Daniel and Zhang, Yan and Choudhary, Jyoti S and Gerstein, Mark and Guigó, Roderic and Hubbard, Tim J P and Kellis, Manolis and Paten, Benedict and Tress, Michael L and Flicek, Paul},
month = jan,
year = {2021},
pages = {D916--D923},
}

Chromatin accessibility (ENCODE)

Histone modification (ENCODE)

CpG methylation (ENCODE)

@article{noauthor_integrated_2012,
title = {An {Integrated} {Encyclopedia} of {DNA} {Elements} in the {Human} {Genome}},
volume = {489},
issn = {0028-0836},
url = {https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3439153/},
doi = {10.1038/nature11247},
number = {7414},
urldate = {2023-05-23},
journal = {Nature},
month = sep,
year = {2012},
pmid = {22955616},
pmcid = {PMC3439153},
pages = {57--74},
}

Enhancer annotation (Fulco et al., Gasperini et al., Avsec et al. )

Enhancers

@article{fulco_activity-by-contact_2019,
title = {Activity-by-contact model of enhancer–promoter regulation from thousands of {CRISPR} perturbations},
volume = {51},
copyright = {2019 The Author(s), under exclusive licence to Springer Nature America, Inc.},
issn = {1546-1718},
url = {https://www.nature.com/articles/s41588-019-0538-0},
doi = {10.1038/s41588-019-0538-0},
language = {en},
number = {12},
urldate = {2023-05-23},
journal = {Nature Genetics},
author = {Fulco, Charles P. and Nasser, Joseph and Jones, Thouis R. and Munson, Glen and Bergman, Drew T. and Subramanian, Vidya and Grossman, Sharon R. and Anyoha, Rockwell and Doughty, Benjamin R. and Patwardhan, Tejal A. and Nguyen, Tung H. and Kane, Michael and Perez, Elizabeth M. and Durand, Neva C. and Lareau, Caleb A. and Stamenova, Elena K. and Aiden, Erez Lieberman and Lander, Eric S. and Engreitz, Jesse M.},
month = dec,
year = {2019},
note = {Number: 12
Publisher: Nature Publishing Group},
keywords = {Epigenetics, Epigenomics, Functional genomics, Gene expression, Gene regulation},
pages = {1664--1669},
}

Enhancers

@article{gasperini_genome-wide_2019,
title = {A {Genome}-wide {Framework} for {Mapping} {Gene} {Regulation} via {Cellular} {Genetic} {Screens}},
volume = {176},
issn = {0092-8674},
url = {https://www.sciencedirect.com/science/article/pii/S009286741831554X},
doi = {10.1016/j.cell.2018.11.029},
language = {en},
number = {1},
urldate = {2023-05-23},
journal = {Cell},
author = {Gasperini, Molly and Hill, Andrew J. and McFaline-Figueroa, José L. and Martin, Beth and Kim, Seungsoo and Zhang, Melissa D. and Jackson, Dana and Leith, Anh and Schreiber, Jacob and Noble, William S. and Trapnell, Cole and Ahituv, Nadav and Shendure, Jay},
month = jan,
year = {2019},
keywords = {CRISPR, CRISPRi, RNA-seq, crisprQTL, eQTL, enhancer, gene regulation, genetic screen, human genetics, single cell},
pages = {377--390.e19},
}

Transcription start sites

@article{avsec_effective_2021,
title = {Effective gene expression prediction from sequence by integrating long-range interactions},
volume = {18},
copyright = {2021 The Author(s)},
issn = {1548-7105},
url = {https://www.nature.com/articles/s41592-021-01252-x},
doi = {10.1038/s41592-021-01252-x},
language = {en},
number = {10},
urldate = {2023-05-23},
journal = {Nature Methods},
author = {Avsec, Žiga and Agarwal, Vikram and Visentin, Daniel and Ledsam, Joseph R. and Grabska-Barwinska, Agnieszka and Taylor, Kyle R. and Assael, Yannis and Jumper, John and Kohli, Pushmeet and Kelley, David R.},
month = oct,
year = {2021},
note = {Number: 10
Publisher: Nature Publishing Group},
keywords = {Gene expression, Machine learning, Software, Transcriptomics},
pages = {1196--1203},
}

Noncoding Variant Effects (Expression) (DeepSEA)

DeepSEA's data was sourced from GRASP and the 1000 Genomes Project, which should also be attributed accordingly.

@article{zhou_predicting_2015,
title = {Predicting effects of noncoding variants with deep learning–based sequence model},
url = {https://www.nature.com/articles/nmeth.3547},
doi = {10.1038/nmeth.3547},
language = {en},
number = {10},
urldate = {2023-06-07},
journal = {Nature Methods},
author = {Zhou, Jian and Troyanskaya, Olga G},
year = {2015},
}

Noncoding variant effects (Disease) (ClinVar)

In case the variant consequences categories are used, Ensembl VEP should be attributed.

@article{10.1093/nar/gkz972,
author = {Landrum, Melissa J and Chitipiralla, Shanmuga and Brown, Garth R and Chen, Chao and Gu, Baoshan and Hart, Jennifer and Hoffman, Douglas and Jang, Wonhee and Kaur, Kuljeet and Liu, Chunlei and Lyoshin, Vitaly and Maddipatla, Zenith and Maiti, Rama and Mitchell, Joseph and O’Leary, Nuala and Riley, George R and Shi, Wenyao and Zhou, George and Schneider, Valerie and Maglott, Donna and Holmes, J Bradley and Kattman, Brandi L},
title = "{ClinVar: improvements to accessing data}",
journal = {Nucleic Acids Research},
volume = {48},
number = {D1},
pages = {D835-D844},
year = {2019},
month = {11},
issn = {0305-1048},
doi = {10.1093/nar/gkz972},
url = {https://doi.org/10.1093/nar/gkz972},
eprint = {https://academic.oup.com/nar/article-pdf/48/D1/D835/31698033/gkz972.pdf},
}

FAQ

How are embeddings upsampled?

Due to tokenization strategies, some models by default return less embedding vectors than their number of input nucleotides. As we still require nucleotide-level input for nucleotide-level prediction tasks, we implement upsampling strategies to match the number of returned embeddings to the number of input nucleotides.

Model Upsampling strategy
DNABert The overlapping k-mer tokenization strategy of DNABert causes some "missing embeddings" at the start and the end of the input sequence, as there is no context to build the k-mer tokens from. For k=3, we repeat the first and the last embedding vectors once. For k=4, we repeat the first once and the last twice. For k=5, we repeat the first and the last twice. For k=6, we repeat the first twice and the last three times.
Nucleotide Transformer Due to 6-mer tokenization, each embedding is repeated 6 times. Remainder tokens are single nucleotides and left as-is.
GENA-LM, DNABERT-2 BPE tokens have variable length. We repeat each embedding vector to the length of the sequence represented by its token.