Skip to content

liefeld/pycogaps

 
 

Repository files navigation

PyCoGAPS

Coordinated Gene Activity in Pattern Sets (CoGAPS) implements a Bayesian MCMC matrix factorization algorithm, GAPS, and links it to gene set statistic methods to infer biological process activity. It can be used to perform sparse matrix factorization on any data, and when this data represents biomolecules, to do gene set analysis.

This package, PyCoGAPS, presents a unified python interface, with a parallel, efficient underlying implementation in C++.

Table of Contents

  1. Installation
  2. Usage
    2.1 Running CoGAPS with Default Parameters
    2.2 Running CoGAPS with Custom Parameters
    2.3 Breaking Down the Return Object from CoGAPS
    2.4 Visualizing Output
    2.5 Running CoGAPS in Parallel
  3. Additional Features of CoGAPS
    3.1 Checkpoint System: Saving/Loading CoGAPS Runs
    3.2 Transposing Data
    3.3 Passing Uncertainty Matrix
    3.4 Distributed CoGAPS
  4. Citing CoGAPS

1. Installation

git clone https://github.com/FertigLab/pycogaps --recursive
cd pycogaps
pip3 install . 
OR
python3 setup.py install

2. Usage

To import the python CoGAPS package:

from PyCoGAPS import *

2.1 Running CoGAPS with Default Parameters

The only required argument to CoGAPS is the path to the data. This can be a .csv, .tsv, .mtx, .h5, or .h5ad file containing the data.

# replace with the path to your data, or use this provided example
path = "./data/GIST.csv" 

# run CoGAPS on your dataset
result = CoGAPS(path)
This is pycogaps version  0.0.1
Running Standard CoGAPS on GIST.csv (1363 genes and 9 samples) with parameters: 

-- Standard Parameters --
nPatterns:  3
nIterations:  1000
seed:  0
sparseOptimization:  False

-- Sparsity Parameters --
alpha: 0.01
maxGibbsMass:  100.0

GapsResult result object with 1363 features and 9 samples
3 patterns were learned
About CoGAPS print status messages

While CoGAPS is running it periodically prints status messages. For example, 20000 of 25000, Atoms: 2932(80), ChiSq: 9728, time: 00:00:29 / 00:01:19. This message tells us that CoGAPS is at iteration 20000 out of 25000 for this phase, and that 29 seconds out of an estimated 1 minute 19 seconds have passed. It also tells us the size of the atomic domain which is a core component of the algorithm but can be ignored for now. Finally, the ChiSq value tells us how closely the A and P matrices reconstruct the original data. In general, we want this value to go down - but it is not a perfect measurment of how well CoGAPS is finding the biological processes contained in the data. CoGAPS also prints a message indicating which phase is currently happening. There are two phases to the algorithm - Equilibration and Sampling.

2.2 Running CoGAPS with Custom Parameters

Most of the time we’ll want to set some parameters before running CoGAPS. Parameters are managed with a CoParams object. This object will store all parameters needed to run CoGAPS and provides a simple interface for viewing and setting the parameter values.

# create a CoParams object
params = CoParams(path)

# set desired parameters
setParam(params, "nPatterns", 7) 
# and/or:
setParams(params, {
            'nIterations': 1500,
            'seed': 42,
            'nPatterns': 7,
        })

result = CoGAPS(path, params)
This is pycogaps version  0.0.1
Running Standard CoGAPS on GIST.csv (1363 genes and 9 samples) with parameters: 

-- Standard Parameters --
nPatterns:  7
nIterations:  1500
seed:  42
sparseOptimization:  False

-- Sparsity Parameters --
alpha: 0.01
maxGibbsMass:  100.0

GapsResult result object with 1363 features and 9 samples
7 patterns were learned
Standard & Sparsity Parameters
  • Standard Parameters

    • nPatterns number of patterns CoGAPS will learn
    • nIterations number of iterations for each phase of the algorithm
    • seed random number generator seed
    • sparseOptimization speeds up performance with sparse data (roughly >80% of data is zero), note this can only be used with the default uncertainty
  • Sparsity Parameters

    • alphaA sparsity parameter for feature matrix
    • alphaP sparsity parameter for sample matrix
    • maxGibbsMassA atomic mass restriction for feature matrix
    • maxGibbsMassP atomic mass restriction for sample matrix
Run Configuration Parameters
  • Run Configuration Parameters (these can be passed in to CoGAPS directly)
    • nThreads maximum number of threads to run on
    • messages T/F for displaying output
    • outputFrequency number of iterations between each output (set to 0 to disable status updates)
    • uncertainty uncertainty matrix - either a matrix or a supported file type
    • checkpointOutFile name of the checkpoint file to create
    • checkpointInterval number of iterations between each checkpoint (set to 0 to disable checkpoints)
    • checkpointInFile if this is provided, CoGAPS runs from the checkpoint contained in this file
    • transposeData T/F for transposing data while reading it in - useful for data that is stored as samples x genes since CoGAPS requires data to be genes x samples
    • workerID if calling CoGAPS in parallel the worker ID can be specified
    • asynchronousUpdates enable asynchronous updating which allows for multi-threaded runs
    • nSnapshots how many snapshots to take in each phase, setting this to 0 disables snapshots
    • snapshotPhase which phase to take snapsjots in e.g. "equilibration", "sampling", "all"
# run config arguments can be passed as an argument to CoGAPS
result = CoGAPS(path, params, messages=False, outputFrequency=250)
Distributed Parameters
  • Distributed Parameters
    • distributed must set to "genome-wide"
    • nSets [distributed parameter] number of sets to break data into
    • cut [distributed parameter] number of branches at which to cut dendrogram used in pattern matching
    • minNS [distributed parameter] minimum of individual set contributions a cluster must contain
    • maxNS [distributed parameter] maximum of individual set contributions a cluster can contain
    • explicitSets [distributed parameter] specify subsets by index or name
    • samplingAnnotation [distributed parameter] specify categories along the rows (cols) to use for weighted sampling
    • samplingWeight [distributed parameter] weights associated with samplingAnnotation
Additional Parameters
  • Additional Parameters
    • subsetIndices set of indices to use from the data
    • subsetDim which dimension (1=rows, 2=cols) to subset
    • geneNames vector of names of genes in data
    • sampleNames vector of names of samples in data
    • fixedPatterns fix either 'A' or 'P' matrix to these values, in the context of distributed CoGAPS, the first phase is skipped and fixedPatterns is used for all sets allowing manual pattern matching, as well as fixed runs of standard CoGAPS
    • whichMatrixFixed either 'A' or 'P', indicating which matrix is fixed
    • takePumpSamples whether or not to take PUMP samples

2.3 Breaking Down the Return Object from CoGAPS

CoGAPS returns a dictionary of the result as two representations: an anndata object and GapsResult object. For simplicity and relevancy, we will only consider the anndata object. CoGAPS stores the lower dimensional representation of the samples (P matrix) in the .var slot and the weight of the features (A matrix) in the .obs slot. The standard deviation across sample points for each matrix are stored in the .uns slots.

# this retrieves the anndata result object
result["anndata"]

alt text

TODO: get mean chi sq, get orig params, get version

2.4 Visualizing Output

The result object can be passed on to the analysis and plotting functions provided in the package.

2.4.1 Default Plot
2.4.2 Residuals Plot
2.4.3 Pattern Markers Plot
2.4.4 Binary Plot
2.4.5 Calculate CoGAPS Statistics
2.4.6 Calculate Gene GSS Statistic
2.4.7 Calculate Gene GS Probability

2.4.1 Default Plot

By default, the plot function displays how the patterns vary across the samples.

# plot result object returned from CoGAPS
plot(result)

alt text

2.4.2 Residuals Plot

plotResiduals calculates residuals and produces a heatmap.

plotResiduals(result)

alt text

2.4.3 Pattern Markers Plot

plotPatternMarkers plots a heatmap of the original data clustered by the pattern markers statistic, which computes the most associated pattern for each gene.

plotPatternMarkers(result, legend_pos=None)

alt text

2.4.4 Binary Plot

binaryA creates a binarized heatmap of the A matrix in which the value is 1 if the value in Amean is greater than threshold * Asd and 0 otherwise.

binaryA(result, threshold=3)

alt text

# plotting clustered binary plot
binaryA(result, threshold=3, cluster=True)

alt text

2.4.5 Calculate CoGAPS Statistics

calcCoGAPSStat calculates a statistic to determine if a pattern is enriched in a a particular set of measurements or samples.

# sets is list of sets of measurements/samples
stats = calcCoGAPSStat(result, sets=['Hs.101174', 'Hs.1012'])
{'twoSidedPValue':               
Pattern1  0.496
Pattern2  0.353
Pattern3  0.289, 
'GSUpreg':                       
Pattern1  0.496
Pattern2  0.647
Pattern3  0.711, 
'GSDownreg':               
Pattern1  0.504
Pattern2  0.353
Pattern3  0.289, 
'GSActEst':               
Pattern1  0.008
Pattern2 -0.294
Pattern3 -0.422}

2.4.6 Calculate Gene GSS Statistic

calcGeneGSStat calculates the probability that a gene listed in a gene set behaves like other genes in the set within the given data set.

stats = calcGeneGSStat(result, GStoGenes=['Hs.101174', 'Hs.1012'], numPerm=1000)
Hs.101174  0.422955
Hs.1012    0.391747

2.4.7 Compute Gene GS Probability

computeGeneGSProb computes the p-value for gene set membership using the CoGAPS-based statistics developed in Fertig et al. (2012). This statistic refines set membership for each candidate gene in a set specified in GSGenes by comparing the inferred activity of that gene to the average activity of the set.

stats = computeGeneGSProb(result, GStoGenes=['Hs.101174', 'Hs.1012'])
Hs.101174  0.617193
Hs.1012    0.887583

2.5 Running CoGAPS in Parallel

Non-Negative Matrix Factorization algorithms typically require long computation times and CoGAPS is no exception. In order to scale CoGAPS up to the size of data sets seen in practice we need to take advantage of modern hardware and parallelize the algorithm.

2.5.1 Multi-Threaded Parallelization

The simplest way to run CoGAPS in parallel is to provide the nThreads argument to CoGAPS. This allows the underlying algorithm to run on multiple threads and has no effect on the mathematics of the algorithm i.e. this is still standard CoGAPS. The precise number of threads to use depends on many things like hardware and data size. The best approach is to play around with different values and see how it effects the estimated time.

result = CoGAPS(path, nThreads=1, nIterations=10000, seed=5)

Note this method relies on CoGAPS being compiled with OpenMP support, use buildReport to check.

print(getBuildReport())

2.5.2 Distributed CoGAPS

For large datasets (greater than a few thousand genes or samples) the multi-threaded parallelization isn’t enough. It is more efficient to break up the data into subsets and perform CoGAPS on each subset in parallel, stitching the results back together at the end (Stein-O’Brien et al. (2017)).

In order to use these extensions, some additional parameters are required. We first need to set CoParam's distributed parameter to be genome-wide using setParam. Next, nSets specifies the number of subsets to break the data set into. cut, minNS, and maxNS control the process of matching patterns across subsets and in general should not be changed from defaults. More information about these parameters can be found in the original papers. These parameters need to be set with a different function, setDistributedParameters, than setParam since they depend on each other. Here we only set nSets (always required), but we have the option to pass the other parameters as well.

# first, set distributed param to "genome-wide"
setParam(params, "distributed", "genome-wide")

# then, set other params such as nSets, cut, minNS, maxNS
params.setDistributedParameters(nSets=3)

# last, call CoGAPS as usual
result = CoGAPS(path, params)

Setting nSets requires balancing available hardware and run time against the size of your data. In general, nSets should be less than or equal to the number of nodes/cores that are available. If that is true, then the more subsets you create, the faster CoGAPS will run - however, some robustness can be lost when the subsets get too small. The general rule of thumb is to set nSets so that each subset has between 1000 and 5000 genes or cells. We will see an example of this on real data in the next two sections.

3. Additional Features of CoGAPS

3.1 Checkpoint System: Saving/Loading CoGAPS Runs

CoGAPS allows the user to save their progress throughout the run, and restart from the latest saved “checkpoint”. This is intended so that if the server crashes in the middle of a long run it doesn’t need to be restarted from the beginning. Set the checkpointInterval parameter to save checkpoints and pass a file name as checkpointInFile to load from a checkpoint.

# enabling checkpoint system, saving CoGAPS run
result1 = CoGAPS(mtx_path, params, checkpointInterval=100, checkpointOutFile="example.out", messages=False)

# loading saved CoGAPS run
result2 = CoGAPS(mtx_path, params, checkpointInFile="example.out", messages=False)

3.2 Transposing Data

If your data is stored as samples x genes, CoGAPS allows you to pass transposeData=True and will automatically read the transpose of your data to get the required genes x samples configuration. In order to transpose data, you must pass transposeData=True as an argument to both CoGAPS() and to CoParams().

# create CoParams object with transposeData argument
params = CoParams(path, transposeData=True)

# run CoGAPS as usual, with transposeData argument
result = CoGAPS(path, transposeData=True)

3.3 Passing Uncertainty Matrix

In addition to providing the data, the user can also specify an uncertainty measurement - the standard deviation of each entry in the data matrix. By default, CoGAPS assumes that the standard deviation matrix is 10% of the data matrix. This is a reasonable heuristic to use, but for specific types of data you may be able to provide better information.

# run CoGAPS with custom uncertainty
result = CoGAPS(path, params, uncertainty=GIST_uncertainty.csv)

3.4 Distributed CoGAPS

3.4.1 Methods of Subsetting Data
3.4.2 Additional Return Information
3.4.3 Manual Pipeline

3.4.1 Methods of Subsetting Data

The default method for subsetting the data is to uniformly break up the rows (cols) of the data. There is an alternative option where the user provides an annotation vector for the rownames (colnames) of the data and gives a weight to each category in the annotation vector. Equal sized subsets are then drawn by sampling all rows (cols) according to the weight of each category.

# sampling with weights - use this example or replace with your data

# list row names to set weights for
names = ['IM00', 'IM02']
# list weight corresponding to the row names
wt = [2, 0.5]
# dictionary format
weight = dict(zip(names, wt))
annotation = ['IM00', 'IM02', 'IM00']

# call this method to set weights
params.setAnnotationWeights(annotation=annotation, weights=weight)

result = CoGAPS(path, params)

Finally, the user can set explicitSets which is a list of character or numeric vectors indicating which names or indices of the data should be put into each set. Make sure to set nSets to the correct value before passing explicitSets.

# running CoGAPS with given subsets - use this example or replace with your data

sets = ['IM00', 'IM02']
setParam(params, "explicitSets", sets)

# call this method to set nSets
params.setDistributedParams(nSets=len(sets))

result = CoGAPS(path, params)

3.4.2 Additional Return Information

When running distributed CoGAPS, some additional metadata is returned that relates to the pattern matching process. This process is how CoGAPS stitches the results from each subset back together.

TODO: once implement unmatched patterns

3.4.3 Manual Pipeline

CoGAPS allows for a custom process for matching the patterns together. If you have a result object from a previous run of Distributed CoGAPS, the unmatched patterns for each subset are found by calling getUnmatchedPatterns. Apply any method you like as long as the result is a matrix with the number of rows equal to the number of samples (genes) and the number of columns is equal to the number of patterns. Then pass the matrix to the fixedPatterns argument along with the original parameters for the GWCoGAPS/scCoGAPS run.

TODO: once implement unmatched patterns

4. Citing CoGAPS

If you use the CoGAPS package for your analysis, please cite Fertig et al. (2010)

If you use the gene set statistic, please cite Ochs et al. (2009)

References

Fertig, Elana J., Jie Ding, Alexander V. Favorov, Giovanni Parmigiani, and Michael F. Ochs. 2010. “CoGAPS: An R/C++ Package to Identify Patterns and Biological Process Activity in Transcriptomic Data.” Bioinformatics 26 (21): 2792–3. https://doi.org/10.1093/bioinformatics/btq503.

Ochs, Michael F., Lori Rink, Chi Tarn, Sarah Mburu, Takahiro Taguchi, Burton Eisenberg, and Andrew K. Godwin. 2009. “Detection of Treatment-Induced Changes in Signaling Pathways in Gastrointestinal Stromal Tumors Using Transcriptomic Data.” Cancer Research 69 (23): 9125–32. https://doi.org/10.1158/0008-5472.CAN-09-1709.

Seung, Sebastian, and Daniel D. Lee. 1999. “Learning the Parts of Objects by Non-Negative Matrix Factorization.” Nature 401 (6755): 788–91. https://doi.org/10.1038/44565.

Stein-O’Brien, Genevieve L., Jacob L. Carey, Wai S. Lee, Michael Considine, Alexander V. Favorov, Emily Flam, Theresa Guo, et al. 2017. “PatternMarkers & Gwcogaps for Novel Data-Driven Biomarkers via Whole Transcriptome Nmf.” Bioinformatics 33 (12): 1892–4. https://doi.org/10.1093/bioinformatics/btx058.

About

No description, website, or topics provided.

Resources

License

Stars

Watchers

Forks

Releases

No releases published

Packages

No packages published

Languages

  • Python 85.0%
  • C++ 13.8%
  • Other 1.2%