Skip to main content
  • Research article
  • Open access
  • Published:

Scalable transcriptomics analysis with Dask: applications in data science and machine learning

Abstract

Background

Gene expression studies are an important tool in biological and biomedical research. The signal carried in expression profiles helps derive signatures for the prediction, diagnosis and prognosis of different diseases. Data science and specifically machine learning have many applications in gene expression analysis. However, as the dimensionality of genomics datasets grows, scalable solutions become necessary.

Methods

In this paper we review the main steps and bottlenecks in machine learning pipelines, as well as the main concepts behind scalable data science including those of concurrent and parallel programming. We discuss the benefits of the Dask framework and how it can be integrated with the Python scientific environment to perform data analysis in computational biology and bioinformatics.

Results

This review illustrates the role of Dask for boosting data science applications in different case studies. Detailed documentation and code on these procedures is made available at https://github.com/martaccmoreno/gexp-ml-dask.

Conclusion

By showing when and how Dask can be used in transcriptomics analysis, this review will serve as an entry point to help genomic data scientists develop more scalable data analysis procedures.

Background

Tissue homeostasis results from an intricate gene regulation network that when disrupted can lead to disease. Gene expression is an intermediate state linking the genome to phenotypic outcomes. Biomedical studies have relied on the analysis of gene expression levels to disease state, progression, outcome and treatment [1,2,3]. Changes in gene expression have helped identify patterns and signatures associated with disease (e.g. community-acquired pneumonia [4] or tuberculosis [5]), improve the understanding of aging conditions [6, 7] and complement genetic information in Mendelian disease diagnosis [8]. Furthermore, analyzing these alterations can facilitate the discovery, development and assessment of novel drug treatments [9,10,11], the optimization of drug application and dosage in cancer therapies [12, 13], or the prediction of response to therapy [3].

Advances in sequencing technologies have produced an unprecedented amount of multi-modal molecular data. This opened the possibility for a thorough and informative interrogation of genotype-phenotype associations on complex diseases, presenting new opportunities for precision medicine. However, finding statistically significant and meaningful associations between molecular data and phenotypic or clinical annotations remains a hard challenge. This difficulty is exacerbated by the high dimensionality of the data and, as is the case with gene expression, a considerable stochastic component.

Machine Learning (ML) seeks to automatically capture statistical associations in data while improving knowledge with additional evidence [14,15,16]. ML is of particular interest in computational biology because it can describe biological phenomena without the need to explicitly model them. Supervised ML builds mathematical models that map the input data, described by several attributes or features, to the corresponding output value or label for each instance or sample. The model can then be used to predict the outcome of unseen or incoming data. Figure 1 shows a general flowchart of a supervised learning (SL) pipeline: (1) train/test data split; (2) model training; (3) model evaluation. Discrete categories are determined through classification tasks (Fig. 1a); when predicting numerical values, the task is called regression (Fig. 1b).

In the context of gene expression analysis, there are several examples of phenotype inference from predictive models. Classification has been used for predicting therapeutic or drug response [17, 18], as well as cancer molecular subtype from gene expression, DNA methylation data, or both [19]. Use cases for regression include the prediction of expression levels of target genes from landmark genes [20] or mutational effects from DNA sequences [21].

While ML provides an adequate solution for statistical association challenges, it often incurs substantial computational demands. Processing power and memory requirements are amplified by the size of the data and the complexity of the models. When building models in a single machine, such as a laptop, it might be too costly or even impossible to load large amounts of data into memory. Moreover, while high-performance computing (HPC) solutions are capable of meeting these requirements, it is not always trivial to fully leverage these resources. Therefore, implementations for scalable computing of ML and other data analytics tasks are required.

There are several frameworks for scalable data analytics available for the widely-used Python programming language. In this review we will focus on a particular framework called Dask [22]. Dask divides data into smaller blocks on which to perform highly parallel computations, thus allowing larger data sets to fit in the memory of single machines. It tightly integrates with existing libraries for Python data analytics and shares a similar interface for implementation, which minimizes the need for code rewrites and facilitates the transition to HPC environments. Here we explore the potential of Dask for scalable ML and data science, applied to bulk and single cell transcriptomics, providing usage recommendations supported by numerous code examples and performance tests.

This paper begins with a review on the need for scalable data analysis in computational biology. We start by describing the standard supervised ML workflow and how it can be adapted for gene expression data analysis. In the following section, we look into the specifics of the Python programming language and how it can be used for ML and data science, discussing its advantages and limitations. We then introduce the general concepts of distributed and parallel computing and enumerate different scalable ML frameworks, with a focus on Dask. We perform several benchmarks and comparisons highlighting the advantages of Dask. Finally, we propose guidelines for the efficient use of Dask for transcriptomics analysis. This review is supported by examples of different tasks, comparative performance tests, and supporting code.

Building gene expression predictive models

Gene expression predictive models are built using supervised learning, which often rely on the data being stored in a structured format, such as tables, see Fig. 1. The labels (categorical or numerical) represent the phenotype or the clinical information to be inferred.

Supervised learning pipelines encompass several key steps [23]. First, gene expression and phenotype data are loaded. At this point, preprocessing can be applied to prepare the data for the remaining steps. The data is then split into training and test sets. ML algorithms learn from the training data to generate a predictive model. In supervised learning, training consists in finding the coefficients, known as model parameters, that provide the best mapping between input features and output labels, as judged by intermediate validation scores. Lastly, the trained model is evaluated on the hold-out test set. The test set is used to estimate the generalization error, i.e. how well the model behaves on unseen data. To ensure the robustness of the generalization error, k-fold cross-validation (CV) [24, 25] is customarily performed during model training. This strategy consists in creating several (k) train/test partitions from the original dataset, repeating the model-building process k times, and averaging out k evaluation scores. If the model reaches an evaluation score that is deemed sufficiently high for a specific application, it can be deployed to work with new incoming data.

For the most part, the development of transcriptomics predictive models follows the traditional approach. However, because gene expression levels obtained from RNA-seq experiments are noisy and have a large amplitude, data preprocessing may have an impact on downstream results. Therefore, feature selection, scaling and normalization are required [26,27,28,29,30,31,32]. Feature selection removes less informative genes, for example those with low variability or low expression levels. It can also select biologically-relevant gene subsets (e.g. protein coding). Scaling is used to attenuate fold differences in expression ranges, typically by shifting the data from a linear to a logarithmic scale. Sample normalization methods account for differences that arise from the sequencing process, like library size, batch effects and gene structure.

Although models are trained automatically, the building process can be controlled by static values defined a priori by the user, known as hyper-parameters [33]. Different hyper-parameter values influence the performance of the final predictive model in different ways. Therefore, hyper-parameter optimization (HPO) can be performed during CV in an attempt to lower the generalization error. If the model reaches an optimized performance according to the evaluation measures, the model is selected and applied to new incoming data. Determining the best set of hyper-parameter values is a computationally expensive combinatorial problem. It requires traversing a multidimensional grid of values, with a model to be trained and tested for each combination. Random search alleviates this issue by sampling a limited number of hyper-parameter combinations [34].

To accurately estimate the performance of an ML algorithm, k-fold CV and HPO can be combined into a strategy known as nested CV. While this provides exhaustive performance estimates, it further exacerbates the computational cost incurred by the two techniques in isolation.

Scientific computing with Python

Python is an interpreted, high-level and general-purpose programming language with a focus on readability [35]. It offers interactive and scripting modes that allow for quick prototyping and deployment of a broad range of scientific applications. Data science and ML are areas where Python’s role has greatly expanded in recent years. The Scientific Python Environment (SPE) is at the core of this expansion. The SPE is based on several highly efficient libraries that support numerical and scientific computation, namely: NumPy [36], for storing and operating over large and multi-dimensional arrays and matrices; Scipy [37] for fundamental routines and algorithms in scientific and technical computation; pandas [38] for data manipulation and analysis; matplotlib [39] for plotting and visualization; and scikit-learn [40] for traditional ML. Python and the SPE have thus become popular tools for data science and ML [23]. However, as the size of the data increases and the tasks become more complex and expensive there is a need for improving program efficiency, namely in terms of execution runtime.

Python concurrent and parallel computation

Parallelism and concurrency are two approaches for improving the efficiency of a program. Their use is highly dependent on the underlying architecture of the central processing unit (CPU). In Python, they can be implemented through native packages such as multiprocessing and multithreading.

A process is an independent instance executed in a processor core or node with dedicated memory space. To speed up computation in CPU-intensive tasks, multiprocessing spawns multiple processes that execute parts of a task in parallel. A thread is run within a process and allows parts of a program to run concurrently when there is a separate flow of execution. Multiple threads can be run in the same process, sharing the same memory space. This reduces the overhead associated with copying and moving data, and making code faster and safer to execute. Multithreading allows concurrency by progressing multiple independent tasks simultaneously.

When multiple threads are launched at the same time, problems may arise if computations are performed out of order. Standard Python implementations address this issue by enforcing the global interpreter lock (GIL) mechanism. The GIL ensures that only a single thread is run at a time. To overcome this important limitation, it is necessary to find solutions for carrying out computations outside the GIL, either by rewriting code in a different language like C or using specialized libraries. In particular, SPE libraries implement several strategies for speeding up computation. For example, NumPy and SciPy can efficiently perform numerical linear algebra operations and bypass the GIL mechanism by leveraging a low-level Basic Linear Algebra Subprograms implementation known as OpenBLAS [41, 42]. SPE libraries also come with the native capability to spawn multiple processes, or jobs, to parallelize computation across several CPU cores. Yet, increasing the number of processes does not always speedup computation. In some cases, excessive parallelization can even be detrimental to overall performance. Therefore, understanding how multiprocessing processes might nest or interact with each other is critical for improving program performance.

Limitations of the scientific Python ecosystem

For all their promise, the advantages of parallelism can be difficult to achieve. To bypass the GIL, multiprocessing has to launch a Python instance containing a full, in-memory copy of the data for each additional CPU core in use. Furthermore, certain ML tasks such as nested CV or hyper-parameter optimization incur heavy computational burdens. Hence, the parallelization of ML pipelines rapidly fills available memory. Furthermore, increases in data size lead to difficulties in data processing and analysis. The limitations of a single machine become apparent when datasets no longer fit comfortably in memory or take too long to load and process. One approach to alleviate these issues would be to share the workload across several machines, but in Python this is not a straightforward task. Data science libraries like pandas or scikit-learn are not designed specifically to operate on distributed systems [43]. It is thus imperative to find computational paradigms that can handle multiple ML models simultaneously built from large or massive datasets and take full advantage of all available CPU cores through efficient parallelization, while offering the possibility to scale beyond a single machine.

Scalable data science

The transformative power of data science is supported by advances in computing capabilities that enable the production, collection, storage and processing of increasingly larger amounts of data. Thus, the computational requirements of large datasets are an important bottleneck in data analysis. This bottleneck can be overcome by improving the quality and/or quantity of computational resources available to a single machine (scaling-up), or spreading computational load across several machines (scaling-out).

Scalable data analytics frameworks can assist in implementing these strategies, ensuring that available resources are fully and expanding computational capabilities in tasks such as ML. For example, when a dataset does not entirely fit in memory, out-of-core computation can be a solution. Out-of-core algorithms are optimized to efficiently fetch and transfer batches of data from disk to memory and to process them in smaller blocks, expanding total available memory.

For several years, the Apache Spark [44, 45] framework has been a popular choice for scalable data analytics. Spark is a unified engine written in Scala for distributed data processing. As a part of the all-in-one Apache ecosystem, Spark interfaces well with other Apache projects and is capable of integrating and parallelizing computation across several languages, namely Java, R, and Python (via the Pyspark library). However, integration with these languages requires additional programming efforts and often incurs serialization costs, as abstract data structures need to be converted between languages.

As such, several frameworks and tools are currently being developed under the Python ecosystem to scale one or several steps of data analysis across several CPUs (and, in some cases, GPUs) while minimizing the need for code rewrites (Table 1). Some frameworks focus on out-of-core parallelization of large tabular datasets, while others provide support for a complete analysis pipeline in a distributed environment. Overall, there is an ongoing effort to develop scalable data science frameworks that provide seamless integration and compatibility with existing SPE code. These frameworks aim to alleviate the burden associated with technical implementation, allowing researchers to focus on scientific questions.

From among the solutions for scalable data science in Python, this paper highlights the Dask framework [22].

Scaling computational biology with Dask

Dask has been applied to a variety of scalable problems in computational biology. It has been used to introduce parallel computation in molecular dynamics analysis and simulations [46,47,48,49], efficiently handle genotype data as arrays [50], assess the reliability of methodologies for the analysis of human brain white matter connections [51], as a component in general neuroimagining pipelines [52], and for powering a workflow in gene regulatory network model exploration [53].

Applications of Dask specific to omics data analysis include scaling the reconstruction of gene regulatory networks from single-cell yeast RNA [54], integrative analysis of multi-omics data [55], inferring gene regulatory networks from large single-cell gene expression datasets [56, 57], analysis of high resolution rRNA sequencing data from multiple amplicons [58], and the study of tissue organization and cellular communications derived from spatial omics data analysis and visualization [59].

Furthermore, Dask plays a supporting role as an underlying component for several data science and machine learning tools used in computational biology. For example, the GPU-accelerated tool RAPIDS [60] builds on top of the Dask framework in order to scale its data preparation and model training steps across multiple GPUs and machines [23]. In this assisting capacity, Dask has made it possible to scale single-cell analysis pipelines to upwards of millions of cells [61]. In large-scale distributed environments, i.e. supercomputers, Dask has been deployed as a workflow manager for GPU-accelerated computation to predict protein structures from genomic sequences [62].

The Dask framework

Dask [22] is a native Python framework that extends the SPE’s capabilities for multi-core and out-of-core computation. This allows Dask to scale beyond a single machine and fit increasingly large datasets into memory. By cloning the APIs of commonly used libraries like NumPy, pandas or scikit-learn, Dask minimizes the changes required to port pre-existing code [36]. The simplicity of Dask greatly reduces the barrier to entry for analysts that are new to distributed and parallel computing. This is especially important in domains such as computational biology and bioinformatics where data analysis pipelines are often developed by scientists without a formal computer science background using the SPE in local workstations.

The Dask framework combines blocked algorithms with task scheduling to achieve parallel and out-of-core computation. The framework is composed of collections, task graphs and schedulers. Collections are data structures that represent and organize the data. Task graphs and schedulers determine how computations are performed (Fig. 2).

Dask implements three collection types specialized in building task graphs for structured and unstructured data: Dask Arrays, Dask Dataframes and Dask Bags. Dask Dataframes are limited to two-dimensional tables, while Dask Arrays can have higher dimensionality. Dask Dataframes split the dataset into multiple partitions along the index. Each partition consists of a pandas Dataframe that can be processed independently, parallelizing the workload. Dask Arrays creates chunks by splitting NumPy arrays along the index and columns. Bags are a more flexible data type that can hold any combination of Python objects. They are particularly useful for working with unstructured data in tasks such as graph or text mining. Because it mimicks the interface of the pandas and NumPy libraries, Dask offers many of the same statistical and analytical functionalities.

In addition, Dask provides the more general Delayed and Futures interfaces that operate as building blocks for implementing custom data structures and algorithms in Dask. Like the aforementioned specialized collections, Dask Delayed executes task graphs lazily as needed, while Future executes functions eagerly and concurrently across multiple cores and machines.

To efficiently handle collections, Dask builds task graphs with blocked algorithms. Blocked algorithms are an approach to out-of-core, distributed computation. Blocked algorithms split data into smaller blocks that are loaded on demand and processed in parallel [63].

A task graph is a directed acyclic graph used to keep track of all tasks and their order. Each node is a function and the edges are objects generated by the preceding function node and used as input by the succeeding node (Fig. 2). Graph evaluation is performed lazily, meaning that task processing is delayed until computation is explicitly called. Due to the great flexibility of blocked algorithms, task graphs can interface with several problem assignment algorithms, such as task scheduling [64].

Task scheduling allows Dask to achieve parallelization by dividing the program into smaller tasks. For instance, when summing three partitions with three numbers each, the scheduler will assign one worker to each partition and process them in parallel. Once this done, the resulting smaller sums are aggregated into a final solution, Fig. 2.

In Dask, there are two groups of task schedulers: single-machine and distributed. Single-machine schedulers leverage local processes or threads, while distributed schedulers operate both locally and across multiple machines. While single-machine schedulers do not require setup and incur in a smaller overhead, distributed schedulers are more sophisticated and offer more features such as a diagnostics dashboard for monitoring performance.

Single-machine schedulers include the threaded scheduler, multiprocessing scheduler and single-threaded synchronous scheduler. The single-threaded scheduler has no parallelism, performing all computations in a single thread, which facilitates debugging. The threaded scheduler leverages a single machine’s entire thread pool, which allows for faster computation times for code that releases the GIL. Lastly, the multiprocessing scheduler provides parallelism by assigning tasks to local processes. This is useful for bypassing the GIL when it cannot be released, as is often the case with pure Python code. However, because inter-process communication is slower than threaded execution, the multiprocessing scheduler does not always reduce execution times.

While single-machine schedulers ship threads or processes to local thread or process pools directly, distributed schedulers launch and interface with computations through clients that assign tasks to workers. In Dask, one worker corresponds to one process, with access to a worker-specific subset of the thread pool. One of the workers is appointed as the master which will coordinate other workers and direct them to execute the individual tasks found in graphs. As tasks are completed, workers become free and are assigned to any remaining tasks until all tasks are executed.

The primary purpose of distributed schedulers is to parallelize computation in clusters. However, clients for interfacing with distributed schedulers can also be initialized locally. This might be desirable for a number of reasons. As previously mentioned, distributed schedulers grants users with access to a diagnostics dashboard which allows them to assess computational performance. Furthermore, distributed schedulers offer improved data locality. Data locality consists in the process of moving computations closer to where data is stored, which is often more efficient than the reverse operation. In such cases, the additional overheard caused by setting up a distributed scheduler locally may prove worthwhile. Lastly, the distributed scheduler can leverage additional worker resources, such as additional memory for the local processing of large datasets or extra CPUs for CPU-intensive computations.

Methods

The source code for processing the data and performing all analyses can be found at: https://github.com/martaccmoreno/gexp-ml-dask.

Assessing phenotype predictive model performance

Using an XGBoost-powered classifier [20] with default hyper-parameters, we compared the performance of the three strategies (Distributed Threads, Distributed Processes and SPE) in a single machine environment for several phenotype prediction tasks.

In all cases, expression matrices comprise n sample rows characterized by f gene feature columns; label vectors have length n. Because all preprocessing steps used Dask, we took advantage of the increase in parallelization whenever possible.

After transcriptome profiling, the BRCA gene expression datasets underwent FPKM normalization. The dataset has 1,205 samples with BRCA molecular subtype information available, profiled across 60,483 genes. Following feature selection of coding genes we obtained a second, smaller feature matrix, in which the number of genes was reduced to 19,564.

The LUAD/LUSC gene expression matrix contained 776 samples and 19,560 genes. These dimensions resulted from filtering for samples of individuals with information on the number of cigarettes smoked per day. Only coding gene features were selected. Gene expression quantification files for this dataset underwent FPKM and UpperQuartile normalizations.

Synthetic read counts for the SYNTH dataset were generated using compcodeR [65], an R package that generates simulated count data. Tweaks in the simulated data distribution were used to mimic different classes. Simulated counts and their respective classes were later subsampled and shuffled with a Python script. The resulting SYNTH dataset has 5,000 samples and 20,000 genes.

Generating datasets with different dimensionalities

For dataset subsampling two approaches were applied: (i) sample-wise subsampling, in which the top 20,000 genes with higher variance were selected as features, and a varying number of samples n (200, 600, and 1,205) was randomly sampled; (ii) feature-wise subsampling, in which the number of samples was fixed as 1,205 (the total), and varying the number of features f was select from among those with the highest variance (1,000, 20,000 and 40,000).

Cross-validation

As of the time of writing, Dask has no official CV support; the proposed solution is to parallelize computation using joblib. However, this only scales CPU usage and does not help with scaling memory. This means that while runtime will be reduced, memory usage will peak similarly as in SPE. We have developed our own custom CV script to use with Dask in tasks involving gene expression data, which can be found in the following repository: https://github.com/martaccmoreno/gexp-ml-dask. This implementation leverages Dask’s out-of-core capabilities for adding disk drive memory to total available memory.

Hyper-parameter optimization

The performance of Dask and SPE in intensive optimization tasks was evaluated by performing an exhaustive grid search on the coding subset of the BRCA dataset (n1,205 \(\times\) f19,564). To that end, a grid containing two values for each of three XGBoost hyper-parameters was transversed as many times as the total number of combinations (2 \(\times\) 2 \(\times\) 2 = 8). Training set performance was evaluated using 5 \(\times\)2 nested CV.

Single-cell RNA-seq preprocessing

To assess how Dask performs and compares against SPE in handling scRNA-seq data, the following tasks were performed:

  1. 1.

    Log-normalization of the data (log-norm);

  2. 2.

    Identification and selection of highly variable gene features (feature selection);

  3. 3.

    Data scaling to shift the distribution to a mean of 0 and standard deviation of 1.

This pipeline was applied in a single machine environment using two frameworks, Dask and SPE, on single-cell transcriptomes sampled from esophagus tissue. The dataset comprised 87,947 samples and 24,245 genes. Smaller dataframes were derived by randomly choosing samples and features without replacement.

Results

To highlight the relevance of the use of scalable data analysis frameworks, we benchmark and compare the performance of the Dask and SPE frameworks in two scenarios: (i) an end-to-end ML pipeline for phenotype prediction from cancer and synthetic bulk transcriptomic data, with and without hyper-parameter optimization; (ii) in the preprocessing single-cell RNA-seq data. All tests were performed on a laptop running Linux Ubuntu 20.04.2 LTS with a 500 GB SSD, 16 GB of RAM and a multi-core processor powered by four cores comprising two threads each, with core frequency of up to 4.6 GHz. or the Dask framework, we have opted to use the Distributed scheduler due to the advantages presented in the previous section. Dask Distributed is deployed using two configurations: (i) Distributed Threads (DT), which spawns a single worker with access to all eight CPU threads in the thread pool; and (ii) Distributed Processes (DP), which spawns four workers, each with access to two threads.

Inferring phenotypes from cancer transcriptomic data

Cancer is a complex and heterogeneous disease driven by genetic alterations which often result in gene expression dysregulation [66, 67]. RNA profiling [26, 68] of tumoral tissues is widely used to uncover cancer gene signatures. Accurate diagnosis is essential to achieve the best clinical outcome [69]. Predictive models of cancer molecular subtypes have been developed from transcriptomic data [70,71,72,73,74], which can assist clinicians deliver specific tumor tailored treatments [75,76,77,78,79], as well as provide further cancer subtype stratification [80,81,82]. Prognostic signatures for assessing patient disease-free survival have been derived with ML from whole-transcriptome data in several tissues [81, 83, 84].

Supervised learning tasks

Transcriptome data from the Cancer Genome Atlas (TCGA) [85] was used to show how phenotypes can be predicted from tumor-specific gene expression levels. Classification and regression models were built with SPE and Dask (Distributed Threads and Distributed Processes) in various ways. The performance of the three approaches is compared in terms of minimum run time, peak memory usage and accuracy for different predictive tasks:

  1. 1

    Classification of five breast cancer molecular subtypes from breast gene expression data sequenced by TCGA (BRCA);

  2. 2

    Prediction of the number of cigarettes smoked per day in individuals diagnosed with lung cancer based on lung gene expression data from TCGA (LUAD/LUSC);

  3. 3

    Binary classification of synthetic samples from read counts generated in silico using different distributions to mimic two distinct classes (SYNTH).

The minimum runtime and peak memory usage values for the three supervised learning tasks are shown in Table 2. Minimum runtime is the shortest execution runtime taken from three repetitions. This metric was chosen since fluctuations in runtime are usually caused by external factors, e.g. other processes running in the background, meaning that the smallest value approximates the true execution time for the code under evaluation. Memory usage reports the highest value recorded during execution.

For the full BRCA dataset, the DT configuration offered gains of at least 11.82% for peak memory usage and 39.4% in runtime compared to DP and SPE. Likewise, DT outperformed the other two methods for the coding subset of the BRCA datset with gains of at least 35.5% and 14.6% for peak memory usage and minimum runtime, as well as the LUAD/LUSC dataset with a minimum improvement in performance of 17.6% and 10.1% for those same metrics. The results for the SYNTH dataset presented a trade off: while DT offered gains of 9.0% for peak memory consumption, DP had a minimum runtime that was 36.2% lower.

Dataset dimensionality

In addition to comparing performance for three datasets, we sought to test and compare the performance of the different strategies when applied to datasets with varying dimensionality, as they may be distinctively affected by a growing number of features (columns) or samples (rows). With subsampling we derived datasets of multiple sizes from the full BRCA dataset, see Fig. 3.

Overall, DT outperformed both DP and SPE in all cases, with the lowest peak memory usage and minimum runtime irrespective of dimensionality.

Intensive optimization tasks

Hyper-parameter optimization (HPO) performance was assessed for the three strategies using the coding subset of the BRCA dataset, see Table 2. DT outperformed the other methods for peak memory usage, with gains of at least 38.6%. Minimum runtime was similar for both Dask configurations, with gains of roughly 36% compared to SPE. All strategies exhibited similarly high levels of accuracy.

Single-cell data analysis

Single-cell RNA sequencing (scRNA-seq) is enabling the study of gene expression at an unprecedented resolution to characterize complex tissues and disease [86]. The constant development of single-cell technologies resulted in an exponential growth in terms of cell profiled [87].

scRNA-seq computational analysis workflows follow a series of well-established steps [88]. Given the count matrices obtained by raw sequencing data alignment, the data is preprocessed before passing through downstream analysis [89]. Given the large amounts of data generated, scalable methods are essential for working with scRNA-seq data.

Here, we show how Dask can scale several tasks (log-scale transformation, feature selection and scaling) commonly performed in the preprocessing of scRNA-seq data. Benchmarks are performed on a dataset sampled from esophagus tissue used to measure ischaemic sensitivity of human tissue at different time points [90]. The performance of DT and DP are compared with SPE relative to the minimum process runtime for different dimensionalities sampled from the complete dataset, see Fig. 4a.

In this scenario, for the lower dimensionality datasets, SPE outperforms both Dask Distributed configurations. However, for larger dimensiontionalities, both configurations surpass SPE and are, in fact, the only viable solution as SPE runs out of memory. Remarkably, the multiprocessing DP approach outperformed DT in all cases. To investigate this difference, we looked into the bottleneck for this simple pipeline: loading times (Fig. 4b).

Given that loading entire datasets into memory was not possible for higher dimensionalities, we tested DT and DP loading times on a subset of the available dimensionalities. As expected, loading comprises a considerable portion of runtime compared to the full pipeline (around 50%). DP always outperformed DT, meaning that even for the smallest dataset (n40k \(\times\) f10k, with file size 1.8 GB), multiprocessing approaches greatly benefited loading times.

Discussion

Getting started with Dask is simple, but knowing when and how to properly deploy it is vital for obtaining the best performance. As the results show, there are some important prerequisites to consider before choosing to scale computation with Dask.

Dataset dimensionality and learning complexity are the two major determinants of the analysis setup. Regarding the size of the dataset, one of the following scenarios can occur: (a) data fits comfortably in memory; (b) data cannot totally fit in memory unless disk drives are leveraged to expand total available memory; and (c) very large datasets that cannot be processed in a single machine. As for learning complexity, computational runtimes of data fitting are proportional to the complexity of the workflow. This step can be expedited with parallelization.

Figure 5 shows different scenarios where SPE and Dask can be applied to surpass limitations and improve performance in ML models trained on datasets of different sizes, with or without intensive hyper-parameter optimization. Note that these guidelines are not strict. For example, if the size of large datasets can be reduced to fit in memory through pre-training processing with Dask, it may be more efficient to switch to SPE for the remainder of the computations.

Dask usage guidelines for transcriptomics analysis

While integrating code with Dask requires minimal rewrites, to optimize performance it is important to consider the specificities of distributed computation explored in this paper, as well as the presented performance benchmarking results. In this section, we give usage guidelines based on our experience from working with transcriptomic data.

Transcriptome data is often presented as a singular tabular file. However, this format does not take full advantage of distributed computation. A naive approach would be to split data into several tabular files, enabling Dask to load and process each file as an independent partition to parallelize computation and reduce memory footprint. However, there are other file formats more suited for distributed computation, such as Parquet [91].

Parquet is a columnar file format that efficiently compresses stored data. It can easily store data as multiple files of fixed size that can be loaded as parallel partitions. Importantly, Parquet stores metadata information, which can greatly enhance Dask’s performance. This especially true of pipelines that include one of several operations that require knowing the index a priori, like sorting. Furthermore, metadata stores the data type of each column, which would otherwise have to be determined through a computationally expensive process of per-column sampling.

Partition size plays a significant role in performance. Ideally, partitions should be small enough to fit into each worker’s memory, but large enough to reduce the overhead caused by each additional partition. Ideal partition size depends on the characteristics and dimensions of each dataset. In our experiments, sizes between 60 to 100 MiB offered a good tradeoff. As several of the data analysis steps, like feature selection, reduce dataset size, distributing data across fewer blocks (repartition) and storing the result in memory (persist) can lead to more efficient task graph execution.

The choice between Dask Distributed Threads or Distributed Processes depends on the size of data, types of execution bottlenecks and proportion of GIL-bound code. Threads should be used to work with numeric collections (e.g. Arrays and Dataframes) that release the GIL. On the other hand, pure Python collections (e.g. Bags) are bound to the GIL and thus typically show gains in performance when leveraging several processes. For example, the data used for building predictive models in our supervised learning tests was small (under 1GB), but because the workflows mostly comprised highly vectorized computations, multithreading had a better overall performance. For the single-cell preprocessing pipeline, multiprocessing exhibited the best performance. This difference was also observed when testing loading times only. Since the file sizes for datasets varied between 1.8GB and 9.5GB, we posit that multiprocessing approaches generally outperform multithreading for larger data sizes and when loading times are an execution bottleneck.

Conclusions

Genomic technology developments have led to an exponential increase in the volume of data collected with multiple molecular assays. The molecular characterization of cohorts of hundreds of individuals (e.g. GTEx [92], TCGA [85], Geuvadis [93]), of diverse cellular characteristics (e.g. ENCODE [94, 95] or the Roadmap Epigenomics Project [96]), and thousands of single cells [88, 97] has been achieved. This has prompted breakthrough advances in many bioinformatics topics, including precision medicine, cancer genetics, population genomics, and developmental molecular biology.

ML appears as a critical tool to map the relationship between the state of molecular entities and phenotypic traits. The magnitude and the high-dimensionality of transcriptomic data often requires considerable computational resources, in the realm of high-performance computing, which are not always available. Thus, alternative approaches for scalable machine learning and data analysis are required.

Dask is highly flexible and versatile and can be used as standalone tool or to support other frameworks. It facilitates scalable data analysis with multi-core and out-of-core computation functionalities. By cloning the API of commonly used scientific Python libraries, Dask makes its adoption rapid and seamless. Although we have focused on so-called shallow learning methods, Dask can also interface with Python Deep Learning libraries [60, 98].

Through several application examples we show that Dask can improve the performance of transcriptomics data analysis and scale computation beyond the usual limits. We foresee that frameworks like Dask will become an essential part of the computational data scientist’s toolkit, alleviating the burden of technical implementation and allowing researchers to concentrate on scientific questions.

Fig. 1
figure 1

Supervised learning for predicting cancer-related phenotypes from gene expression data. a Classification identifies target labels, including cancer subtypes; b regression can predict progression and outcome measures, such as disease-free intervals. After the data is partitioned into training and test sets, a ML algorithm is fit on the training data. The model is evaluated using hold-out test data

Fig. 2
figure 2

Main components of the Dask framework. Collections are processed by task graphs, which are executed by schedulers. In a task graph the nodes represent functions (f) and edges are Python objects (o)

Fig. 3
figure 3

Performance comparison of the three strategies for the subsampled BRCA datasets. Peak memory, minimum runtime and classification accuracy were measured. Differences in accuracy resulted from the algorithmic implementation specificities of Dask and SPE. The data sizes shown in MB correspond to uncompressed tabular files

Fig. 4
figure 4

Runtime comparison between the Dask and SPE frameworks in A the preprocessing and B the full loading of scRNA-seq data. Datasets were subsampled for different dimensions. In A, both Dask Distributed configurations (Threads and Processes) partially load the data, processing dataset partitions; in B, the entire dataset is loaded. Asterisks represent instances when programs ran out of memory. n is the number of rows and f the number of features. The file sizes shown in GB correspond to uncompressed tabular files

Fig. 5
figure 5

Proposed usage of SPE and Dask for different scenarios of model complexity and data size. When Dask and SPE are combined, Dask is used for preprocessing and SPE for data analysis and ML

Table 1 Examples of Python tools and frameworks for scalable data science
Table 2 Summary of memory and runtime performance for the three strategies on different datasets

Availability of data and materials

All source code used to generate the results used in this paper, including steps on how to obtain the data from public repositories, can be found at: https://github.com/martaccmoreno/gexp-ml-dask.

Abbreviations

CPU:

Central processing unit

CV:

Cross-validation

DP:

Distributed processes

DT:

Distributed threads

GIL:

Global interpreter lock

HPC:

High-performance computing

HPO:

Hyper-parameter optimization

ML:

Machine learning

SPE:

Scientific Python environment

scRNA-seq:

Single-cell RNA sequencing

SL:

Supervised learning

TCGA:

The Cancer Genome Atlas

References

  1. Byron SA, Keuren-Jensen KRV, Engelthaler DM, et al. Translating RNA sequencing into clinical diagnostics: opportunities and challenges. Nat Rev Genet. 2016;17(5):257–71. https://doi.org/10.1038/nrg.2016.10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Casamassimi A, Federico A, Rienzo M, Esposito S, Ciccodicola A. Transcriptome profiling in human diseases: new advances and perspectives. Int J Mol Sci. 2017;18(8):1652. https://doi.org/10.3390/ijms18081652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sammut S-J, Crispin-Ortuzar M, Chin S-F, Provenzano E, Bardwell HA, Ma W, Cope W, Dariush A, Dawson S-J, Abraham JE, et al. Multi-omic machine learning predictor of breast cancer therapy response. Nature. 2021;601:1–10.

    Google Scholar 

  4. Scicluna BP, Klouwenberg PMCK, van Vught LA, et al. A molecular biomarker to diagnose community-acquired pneumonia on intensive care unit admission. Am J Respir Crit Care Med. 2015;192(7):826–35. https://doi.org/10.1164/rccm.201502-0355oc.

    Article  CAS  PubMed  Google Scholar 

  5. Sweeney TE, Braviak L, Tato CM, et al. Genome-wide expression for diagnosis of pulmonary tuberculosis: a multicohort analysis. Lancet Respir Med. 2016;4(3):213–24. https://doi.org/10.1016/s2213-2600(16)00048-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Glass D, Viñuela A, Davies MN, et al. Gene expression changes with age in skin, adipose tissue, blood and brain. Genome Biol. 2013;14(7):1–12. https://doi.org/10.1186/gb-2013-14-7-r75.

    Article  CAS  Google Scholar 

  7. Fleischer JG, Schulte R, Tsai HH, et al. Predicting age from the transcriptome of human dermal fibroblasts. Genome Biol. 2018;19(1):1–8. https://doi.org/10.1186/s13059-018-1599-6.

    Article  CAS  Google Scholar 

  8. Cummings BB, Marshall JL, Tukiainen T, et al. Improving genetic diagnosis in Mendelian disease with transcriptome sequencing. Sci Transl Med. 2017;9(386):5209. https://doi.org/10.1126/scitranslmed.aal5209.

    Article  Google Scholar 

  9. Vamathevan J, Clark D, Czodrowski P, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463–77. https://doi.org/10.1038/s41573-019-0024-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cliff JM, Lee J-S, Constantinou N, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J Infect Dis. 2013;207(1):18–29. https://doi.org/10.1093/infdis/jis499.

    Article  CAS  PubMed  Google Scholar 

  11. Murray PG, Stevens A, Leonibus CD, et al. Transcriptomics and machine learning predict diagnosis and severity of growth hormone deficiency. JCI Insight. 2018. https://doi.org/10.1172/jci.insight.93247.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Huang C, Mezencev R, McDonald JF, et al. Open source machine-learning algorithms for the prediction of optimal cancer drug therapies. PLoS ONE. 2017;12(10):0186906. https://doi.org/10.1371/journal.pone.0186906.

    Article  CAS  Google Scholar 

  13. Sakellaropoulos T, Vougas K, Narang S, et al. A deep learning framework for predicting response to therapy in cancer. Cell Rep. 2019;29(11):3367–73. https://doi.org/10.1016/j.celrep.2019.11.017.

    Article  CAS  PubMed  Google Scholar 

  14. Deo RC. Machine learning in medicine. Circulation. 2015;132(20):1920–30. https://doi.org/10.1161/circulationaha.115.001593.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Libbrecht MW, Noble WS. Machine learning applications in genetics and genomics. Nat Rev Genet. 2015;16(6):321–32. https://doi.org/10.1038/nrg3920.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jordan MI, Mitchell TM. Machine learning: trends, perspectives, and prospects. Science. 2015;349(6245):255–60. https://doi.org/10.1126/science.aaa8415.

    Article  CAS  PubMed  Google Scholar 

  17. Kang T, Ding W, Zhang L, et al. A biological network-based regularized artificial neural network model for robust phenotype prediction from gene expression data. BMC Bioinform. 2017;18(1):1–11. https://doi.org/10.1186/s12859-017-1984-2.

    Article  CAS  Google Scholar 

  18. Aliper A, Plis S, Artemov A, et al. Deep learning applications for predicting pharmacological properties of drugs and drug repurposing using transcriptomic data. Mol Pharm. 2016;13(7):2524–30. https://doi.org/10.1021/acs.molpharmaceut.6b00248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. List M, Hauschild A-C, Tan Q, et al. Classification of breast cancer subtypes by combining gene expression and DNA methylation data. J Integr Bioinform. 2014;11(2):1–14. https://doi.org/10.1515/jib-2014-236.

    Article  Google Scholar 

  20. Chen T, Guestrin C. XGBoost: A scalable tree boosting system. In: Krishnapuram B, Shah M (editors) Proceedings of the 22nd ACM SIGKDD international conference on knowledge discovery and data mining. ACM, San Francisco, California; 2016. p. 785–94. https://doi.org/10.1145/2939672.2939785. arXiv:1603.02754.

  21. Zhou J, Theesfeld CL, Yao K, et al. Deep learning sequence-based ab initio prediction of variant effects on expression and disease risk. Nat Genet. 2018;50(8):1171–9. https://doi.org/10.1038/s41588-018-0160-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rocklin M. Dask: parallel computation with blocked algorithms and task scheduling. In: Proceedings of the 14th Python in science conference, vol 130. SciPy, Austin, Texas; 2015. p. 136. https://doi.org/10.25080/majora-7b98e3ed-013.

  23. Raschka S, Patterson J, Nolet C. Machine learning in Python: main developments and technology trends in data science, machine learning, and artificial intelligence. Information. 2020;11(4):193. https://doi.org/10.3390/info11040193.

    Article  Google Scholar 

  24. Stone M. Cross-validatory choice and assessment of statistical predictions. J R Stat Soc Ser B (Methodol). 1974;36(2):111–33. https://doi.org/10.1111/j.2517-6161.1974.tb00994.x.

    Article  Google Scholar 

  25. Kohavi R. A study of cross-validation and bootstrap for accuracy estimation and model selection. In: International Joint Conference on Artificial Intelligence, vol. 14. Montreal, Quebec, Canada; 1995. pp. 1137–1143.

  26. Mortazavi A, Williams BA, McCue K, et al. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5(7):621–8. https://doi.org/10.1038/nmeth.1226.

    Article  CAS  PubMed  Google Scholar 

  27. Garber M, Grabherr MG, Guttman M, et al. Computational methods for transcriptome annotation and quantification using RNA-seq. Nat Methods. 2011;8(6):469–77. https://doi.org/10.1038/nmeth.1613.

    Article  CAS  PubMed  Google Scholar 

  28. Zyprych-Walczak J, Szabelska A, Handschuh L, et al. The impact of normalization methods on RNA-seq data analysis. Biomed Res Int. 2015;2015:1–10. https://doi.org/10.1155/2015/621690.

    Article  CAS  Google Scholar 

  29. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 2010;11(3):1–9. https://doi.org/10.1186/gb-2010-11-3-r25.

    Article  CAS  Google Scholar 

  30. Bullard JH, Purdom E, Hansen KD, et al. Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinform. 2010;11(1):1–13. https://doi.org/10.1186/1471-2105-11-94.

    Article  CAS  Google Scholar 

  31. Anders S, Huber W. Differential expression analysis for sequence count data. Nat Preced. 2010. https://doi.org/10.1038/npre.2010.4282.1.

    Article  Google Scholar 

  32. Evans C, Hardin J, Stoebel DM. Selecting between-sample RNA-Seq normalization methods from the perspective of their assumptions. Brief Bioinform. 2018;19(5):776–92. https://doi.org/10.1093/bib/bbx008.

    Article  CAS  PubMed  Google Scholar 

  33. Claesen M, Moor BD. Hyperparameter search in machine learning. arXiv preprint, 2015.

  34. Bergstra J, Bengio Y. Random search for hyper-parameter optimization. J Mach Learn Res. 2012;13(2):281–305.

    Google Scholar 

  35. Perez F, Granger BE, Hunter JD. Python: an ecosystem for scientific computing. Comput Sci Eng. 2010;13(2):13–21. https://doi.org/10.1109/mcse.2010.119.

    Article  Google Scholar 

  36. Harris CR, Millman KJ, van der Walt SJ, et al. Array programming with NumPy. Nature. 2020;585(7825):357–62. https://doi.org/10.1038/s41586-020-2649-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Virtanen P, Gommers R, Oliphant TE, et al. SciPy 1.0:fundamental algorithms for scientific computing in Python. Nat Methods. 2020;17(3):261–72. https://doi.org/10.1038/s41592-019-0686-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. McKinney W. Data structures for statistical computing in python. In: Jones E, Millman J (editors) Proceedings of the 9th Python in science, vol 445. SciPy, Austin, Texas; 2010. p. 51–6. https://doi.org/10.25080/majora-92bf1922-00a.

  39. Hunter JD. Matplotlib: a 2D graphics environment. Comput Sci Engi. 2007;9(3):90–5. https://doi.org/10.1109/mcse.2007.55.

    Article  Google Scholar 

  40. Pedregosa F, Varoquaux G, Gramfort A, et al. Scikit-learn: machine learning in Python. J Mach Learn Res. 2011;12:2825–30.

    Google Scholar 

  41. Xianyi Z, Qian W, Yunquan Z. Model-driven level 3 BLAS performance optimization on Loongson 3A Processor. In: Tang X, Xu C-Z (editors) 2012 IEEE 18th international conference on parallel and distributed systems. IEEE, Washington, DC; 2012. p. 684–91. https://doi.org/10.1109/icpads.2012.97.

  42. Wang Q, Zhang X, Zhang Y, et al. AUGEM: automatically generate high performance dense linear algebra kernels on x86 CPUs. In: Supinski BRd (editor) Proceedings of the international conference on high performance computing, networking, storage and analysis. ACM, New York; 2013. p. 1–12. https://doi.org/10.1145/2503210.2503219.

  43. Daniel JC. Data science at scale with Python and Dask. 1st ed. Shelter Island: Manning Publications; 2019.

    Google Scholar 

  44. Zaharia M, Chowdhury M, Das T, Dave A, Ma J, Mccauley M, Franklin M, Shenker S, Stoica I. Fast and interactive analytics over Hadoop data with spark. Usenix Login. 2012;37(4):45–51.

    Google Scholar 

  45. Zaharia M, Xin RS, Wendell P, Das T, Armbrust M, Dave A, Meng X, Rosen J, Venkataraman S, Franklin MJ, et al. Apache spark: a unified engine for big data processing. Commun ACM. 2016;59(11):56–65.

    Article  Google Scholar 

  46. Dotson David L, Seyler Sean L, Linke Max, et al. datreant: persistent, Pythonic trees for heterogeneous data. In: Benthall S, Rostrup S (editors) Proceedings of the 15th Python in science conference; 2016. p. 51–6 . https://doi.org/10.25080/Majora-629e541a-007.

  47. Khoshlessan M, Paraskevakos I, Jha S, et al. Parallel analysis in MDAalysis using the Dask parallel computing library. In: Ramachandran P, Rey S (editors) Proceedings of the 16th Python in science conference. SciPy, Austin, Texas; 2017. p. 64–72. https://doi.org/10.25080/shinma-7f4c6e7-00a.

  48. Paraskevakos I, Luckow A, Khoshlessan M, et al. Task-parallel analysis of molecular dynamics trajectories. In: Malony AD (editor) Proceedings of the 47th international conference on parallel processing. ACM, Eugene, Oregon; 2018. p. 1–10. https://doi.org/10.1145/3225058.3225128.

  49. Smith P, Lorenz CD. LiPyphilic: a Python toolkit for the analysis of lipid membrane simulations. J Chem Theory Comput. 2021;17(9):5907–19. https://doi.org/10.1021/acs.jctc.1c00447.

    Article  CAS  PubMed  Google Scholar 

  50. Taylor-Weiner A, Aguet F, Haradhvala NJ, et al. Scaling computational genomics to millions of individuals with GPUs. Genome Biol. 2019;20(1):1–5. https://doi.org/10.1186/s13059-019-1836-7.

    Article  Google Scholar 

  51. Kruper J, Yeatman JD, Richie-Halford A, et al. Evaluating the reliability of human brain white matter tractometry. bioarxiv; 2021. https://doi.org/10.1101/2021.02.24.432740.

  52. Dugre M, Hayot-Sasson V, Glatard T. A performance comparison of Dask and Apache spark for data-intensive neuroimaging pipelines. In: Taylor IJ (editor) 2019 IEEE/ACM workflows in support of large-scale science (WORKS). IEEE, Denver, Colorado; 2019. p. 40–9. https://doi.org/10.1109/works49585.2019.00010.

  53. Wrede F, Hellander A. Smart computational exploration of stochastic gene regulatory network models using human-in-the-loop semi-supervised learning. Bioinformatics. 2019;35(24):5199–206. https://doi.org/10.1093/bioinformatics/btz420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Jackson CA, Castro DM, Saldi G-A, et al. Gene regulatory network reconstruction using single-cell RNA sequencing of barcoded genotypes in diverse environments. eLife. 2020;9:51254. https://doi.org/10.7554/elife.51254.

    Article  CAS  Google Scholar 

  55. Tran NC, Gao JX. OpenOmics: a bioinformatics API to integrate multi-omics datasets and interface with public databases. J Open Source Softw. 2021;6(61):3249. https://doi.org/10.21105/joss.03249.

    Article  Google Scholar 

  56. Moerman T, Santos SA, González-Blas CB, et al. GRNBoost2 and Arboreto: efficient and scalable inference of gene regulatory networks. Bioinformatics. 2019;35(12):2159–61. https://doi.org/10.1093/bioinformatics/bty916.

    Article  CAS  PubMed  Google Scholar 

  57. Gibbs CS, Jackson CA, Saldi G-A, et al. High performance single-cell gene regulatory network inference at scale: the Inferelator 3.0. bioRxiv; 2021. https://doi.org/10.1101/2021.05.03.442499.

  58. Debelius JW, Robeson M, Hugerth LW, et al. A comparison of approaches to scaffolding multiple regions along the 16S rRNA gene for improved resolution. bioRxiv; 2021. https://doi.org/10.1101/2021.03.23.436606.

  59. Palla G, Spitzer H, Klein M, et al. Squidpy: a scalable framework for spatial omics analysis. Nat Methods. 2022;19(2):171–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. RD Team. RAPIDS: collection of libraries for end to end GPU data science. Santa Clara: NVIDIA; 2018.

    Google Scholar 

  61. Nolet C, Lal A, Ilango R, et al. Accelerating single-cell genomic analysis with gpus. bioRxiv; 2022.

  62. Gao M, Coletti M, Davidson RB, et al. Proteome-scale deployment of protein structure prediction workflows on the summit supercomputer. arXiv preprint arXiv:2201.10024, 2022.

  63. Lam MD, Rothberg EE, Wolf ME. The cache performance and optimizations of blocked algorithms. ACM SIGOPS Oper Syst Rev. 1991;25(Special Issue):63–74. https://doi.org/10.1145/106973.106981.

    Article  Google Scholar 

  64. El-Rewini H, Ali HH, Lewis T. Task scheduling in multiprocessing systems. Computer. 1995;28(12):27–37. https://doi.org/10.1109/2.476197.

    Article  Google Scholar 

  65. Soneson C. compcoder-an r package for benchmarking differential expression methods for RNA-seq data. Bioinformatics. 2014;30(17):2517–8.

    Article  CAS  PubMed  Google Scholar 

  66. Burrell RA, McGranahan N, Bartek J, et al. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–45. https://doi.org/10.1038/nature12625.

    Article  CAS  PubMed  Google Scholar 

  67. Bradner JE, Hnisz D, Young RA. Transcriptional addiction in cancer. Cell. 2017;168(4):629–43. https://doi.org/10.1016/j.cell.2016.12.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Stark R, Grzelak M, Hadfield J. RNA sequencing: the teenage years. Nat Rev Genet. 2019;20(11):631–56. https://doi.org/10.1038/s41576-019-0150-2.

    Article  CAS  PubMed  Google Scholar 

  69. Miller KD, Nogueira L, Mariotto AB, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 2019;69(5):363–85. https://doi.org/10.3322/caac.21565.

    Article  PubMed  Google Scholar 

  70. Nguyen DV, Rocke DM. Tumor classification by partial least squares using microarray gene expression data. Bioinformatics. 2002;18(1):39–50. https://doi.org/10.1093/bioinformatics/18.1.39.

    Article  CAS  PubMed  Google Scholar 

  71. Li Y, Kang K, Krahn JM, et al. A comprehensive genomic pan-cancer classification using The Cancer Genome Atlas gene expression data. BMC Genom. 2017;18(1):1–13. https://doi.org/10.1186/s12864-017-3906-0.

    Article  CAS  Google Scholar 

  72. Kunz M, Löffler-Wirth H, Dannemann M, et al. RNA-seq analysis identifies different transcriptomic types and developmental trajectories of primary melanomas. Oncogene. 2018;37(47):6136–51. https://doi.org/10.1038/s41388-018-0385-y.

    Article  CAS  PubMed  Google Scholar 

  73. Kim S-K, Kim H-J, Park J-L, et al. Identification of a molecular signature of prognostic subtypes in diffuse-type gastric cancer. Gastric Cancer. 2020;23(3):473–82. https://doi.org/10.1007/s10120-019-01029-4.

    Article  CAS  PubMed  Google Scholar 

  74. Mostavi M, Chiu Y-C, Huang Y, et al. Convolutional neural network models for cancer type prediction based on gene expression. BMC Med Genom. 2020;13(5):1–13. https://doi.org/10.1186/s12920-020-0677-2.

    Article  CAS  Google Scholar 

  75. Khan J, Wei JS, Ringnér M, et al. Classification and diagnostic prediction of cancers using gene expression profiling and artificial neural networks. Nat Med. 2001;7(6):673–9. https://doi.org/10.1038/89044.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Fakoor R, Ladhak F, Nazi A, et al. Using deep learning to enhance cancer diagnosis and classication. In: Proceedings of the international conference on machine learning, vol 28. ACM, New York; 2013. p. 3937–3949.

  77. Iqbal J, Wright G, Wang C, et al. Gene expression signatures delineate biological and prognostic subgroups in peripheral T-cell lymphoma. Blood J Am Soc Hematol. 2014;123(19):2915–23. https://doi.org/10.1182/blood-2013-11-536359.

    Article  CAS  Google Scholar 

  78. Gerami P, Cook RW, Russell MC, et al. Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J Am Acad Dermatol. 2015;72(5):780–5. https://doi.org/10.1016/j.jaad.2015.01.009.

    Article  CAS  PubMed  Google Scholar 

  79. Allen EMV, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG, Goldinger SM, Utikal J, Hassel JC, Weide B, Kaehler KC, Loquai C, Mohr P, Gutzmer R, Dummer R, Gabriel S, Wu CJ, Schadendorf D, Garraway LA. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350(6257):207–11. https://doi.org/10.1126/science.aad0095.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Podolsky MD, Barchuk AA, Kuznetcov VI, et al. Evaluation of machine learning algorithm utilization for lung cancer classification based on gene expression levels. Asian Pac J Cancer Prev. 2016;17(2):835–8. https://doi.org/10.7314/apjcp.2016.17.2.835.

    Article  PubMed  Google Scholar 

  81. Wong N, Khwaja SS, Baker CM, et al. Prognostic micro RNA signatures derived from The Cancer Genome Atlas for head and neck squamous cell carcinomas. Cancer Med. 2016;5(7):1619–28. https://doi.org/10.1002/cam4.718.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sinkala M, Mulder N, Martin D. Machine learning and network analyses reveal disease subtypes of pancreatic cancer and their molecular characteristics. Sci Rep. 2020;10(1):1–14. https://doi.org/10.1038/s41598-020-58290-2.

    Article  CAS  Google Scholar 

  83. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med. 2015;373(21):2005–14. https://doi.org/10.1056/nejmoa1510764.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lim H-Y, Sohn I, Deng S, et al. Prediction of disease-free survival in hepatocellular carcinoma by gene expression profiling. Ann Surg Oncol. 2013;20(12):3747–53. https://doi.org/10.1245/s10434-013-3070-y.

    Article  PubMed  Google Scholar 

  85. Tomczak K, Czerwińska P, Wiznerowicz M. Review The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Współczesna Onkologia. 2015;19(1A):68. https://doi.org/10.5114/wo.2014.47136.

    Article  Google Scholar 

  86. Efremova M, Vento-Tormo R, Park J-E, et al. Immunology in the era of single-cell technologies. Annu Rev Immunol. 2020;38:727–57.

    Article  CAS  PubMed  Google Scholar 

  87. Svensson V, Natarajan KN, Ly L-H, et al. Power analysis of single-cell RNA-sequencing experiments. Nat Methods. 2017;14(4):381–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol. 2019;15(6):8746. https://doi.org/10.15252/msb.20188746.

    Article  Google Scholar 

  89. Andrews TS, Kiselev VY, McCarthy D, et al. Tutorial: guidelines for the computational analysis of single-cell RNA sequencing data. Nat Protoc. 2021;16(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  90. Madissoon E, Wilbrey-Clark A, Miragaia R, et al. scrna-seq assessment of the human lung, spleen, and esophagus tissue stability after cold preservation. Genome Biol. 2020;21(1):1–16.

    Article  CAS  Google Scholar 

  91. Vohra D. Apache parquet. In: Practical Hadoop ecosystem. Berkeley, CA: Apress; 2016. p. 325–35.

  92. Lonsdale J, Thomas J, Salvatore M, et al. The genotype-tissue expression (GTEx) project. Nat Genet. 2013;45(6):580–5. https://doi.org/10.1038/ng.2653.

    Article  CAS  Google Scholar 

  93. Lappalainen T, Sammeth M, Friedländer MR, et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature. 2013;501(7468):506–11. https://doi.org/10.1038/nature12531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. https://doi.org/10.1038/nature11247.

    Article  CAS  Google Scholar 

  95. Davis CA, Hitz BC, Sloan CA, et al. The encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46(D1):794–801. https://doi.org/10.1093/nar/gkx1081.

    Article  CAS  Google Scholar 

  96. Kundaje A, Meuleman W, Ernst J, et al. Integrative analysis of 111 reference human epigenomes. Nature. 2015;518(7539):317–30. https://doi.org/10.1038/nature14248.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018;50(8):1–14. https://doi.org/10.1038/s12276-018-0071-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Holt J, Sievert S. Training machine learning models faster with dask. In: SciPy conferences; 2021

  99. Petersohn D, Macke S, Xin D, et al. Towards scalable dataframe systems. VLDB Endow. 2020;13(12):2033–46. https://doi.org/10.14778/3407790.3407807.

    Article  Google Scholar 

  100. Petersohn D, Tang D, Durrani R, et al. Flexible rule-based decomposition and metadata independence in modin: a parallel dataframe system. Proc VLDB Endow. 2021;15(3):739–51. https://doi.org/10.14778/3494124.3494152.

    Article  Google Scholar 

  101. Moritz P, Nishihara R, Wang S, et al. Ray: a distributed framework for emerging AI applications. In: 13th USENIX symposium on operating systems design and implementation (OSDI 18). USENIX Association, Carlsbad, CA; 2018. p. 561–577.

  102. Totoni E, Hassan WU, Anderson TA, et al. HiFrames: high performance data frames in a scripting language; 2017. arXiv:1704.02341

  103. Breddels MA, Veljanoski J. Vaex: big data exploration in the era of Gaia. Astron Astrophys. 2018;618:13. https://doi.org/10.1051/0004-6361/201732493.

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank all the Sysadmins at MACC HPC center for their assistance. The authors acknowledge Minho Advanced Computing Center for providing HPC resources that have contributed to the research results reported within this paper.

Funding

This work is financed by National Funds through the Portuguese funding agency, FCT - Fundação para a Ciência e a Tecnologia, within project LA/P/0063/2020, the Portuguese National Network for Advanced Computing to PGF for the Grant CPCA/A2/2640/2020, and the Portuguese Foundation for Science and Technology to MM for the Ph.D. scholarship (reference SFRH/BD/145707/2019). No funding body played any role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

PGF and MM designed the study. MM developed the code with initial contributions from PGF. RV assisted with state-of-the-art revision. MM and PGF wrote and revised the manuscript. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Pedro G. Ferreira.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

PGF is a partner of company Bioinf2Bio. The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moreno, M., Vilaça, R. & Ferreira, P.G. Scalable transcriptomics analysis with Dask: applications in data science and machine learning. BMC Bioinformatics 23, 514 (2022). https://doi.org/10.1186/s12859-022-05065-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12859-022-05065-3

Keywords