-
Notifications
You must be signed in to change notification settings - Fork 1
/
Copy pathWorkflow.bib
1555 lines (1526 loc) · 124 KB
/
Workflow.bib
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
126
127
128
129
130
131
132
133
134
135
136
137
138
139
140
141
142
143
144
145
146
147
148
149
150
151
152
153
154
155
156
157
158
159
160
161
162
163
164
165
166
167
168
169
170
171
172
173
174
175
176
177
178
179
180
181
182
183
184
185
186
187
188
189
190
191
192
193
194
195
196
197
198
199
200
201
202
203
204
205
206
207
208
209
210
211
212
213
214
215
216
217
218
219
220
221
222
223
224
225
226
227
228
229
230
231
232
233
234
235
236
237
238
239
240
241
242
243
244
245
246
247
248
249
250
251
252
253
254
255
256
257
258
259
260
261
262
263
264
265
266
267
268
269
270
271
272
273
274
275
276
277
278
279
280
281
282
283
284
285
286
287
288
289
290
291
292
293
294
295
296
297
298
299
300
301
302
303
304
305
306
307
308
309
310
311
312
313
314
315
316
317
318
319
320
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
344
345
346
347
348
349
350
351
352
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423
424
425
426
427
428
429
430
431
432
433
434
435
436
437
438
439
440
441
442
443
444
445
446
447
448
449
450
451
452
453
454
455
456
457
458
459
460
461
462
463
464
465
466
467
468
469
470
471
472
473
474
475
476
477
478
479
480
481
482
483
484
485
486
487
488
489
490
491
492
493
494
495
496
497
498
499
500
501
502
503
504
505
506
507
508
509
510
511
512
513
514
515
516
517
518
519
520
521
522
523
524
525
526
527
528
529
530
531
532
533
534
535
536
537
538
539
540
541
542
543
544
545
546
547
548
549
550
551
552
553
554
555
556
557
558
559
560
561
562
563
564
565
566
567
568
569
570
571
572
573
574
575
576
577
578
579
580
581
582
583
584
585
586
587
588
589
590
591
592
593
594
595
596
597
598
599
600
601
602
603
604
605
606
607
608
609
610
611
612
613
614
615
616
617
618
619
620
621
622
623
624
625
626
627
628
629
630
631
632
633
634
635
636
637
638
639
640
641
642
643
644
645
646
647
648
649
650
651
652
653
654
655
656
657
658
659
660
661
662
663
664
665
666
667
668
669
670
671
672
673
674
675
676
677
678
679
680
681
682
683
684
685
686
687
688
689
690
691
692
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
737
738
739
740
741
742
743
744
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
771
772
773
774
775
776
777
778
779
780
781
782
783
784
785
786
787
788
789
790
791
792
793
794
795
796
797
798
799
800
801
802
803
804
805
806
807
808
809
810
811
812
813
814
815
816
817
818
819
820
821
822
823
824
825
826
827
828
829
830
831
832
833
834
835
836
837
838
839
840
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
866
867
868
869
870
871
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
901
902
903
904
905
906
907
908
909
910
911
912
913
914
915
916
917
918
919
920
921
922
923
924
925
926
927
928
929
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
952
953
954
955
956
957
958
959
960
961
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
Automatically generated by Mendeley Desktop 1.17.13
Any changes to this file will be lost if it is regenerated by Mendeley.
BibTeX export options can be customized via Preferences -> BibTeX in Mendeley Desktop
@article{McCarthy2017,
abstract = {Motivation: Single-cell RNA sequencing (scRNA-seq) is increasingly used to study gene expression at the level of individual cells. However, preparing raw sequence data for further analysis is not a straightforward process. Biases, artifacts and other sources of unwanted variation are present in the data, requiring substantial time and effort to be spent on pre-processing, quality control (QC) and normalization. Results: We have developed the R/Bioconductor package scater to facilitate rigorous pre-processing, quality control, normalization and visualization of scRNA-seq data. The package provides a conveni-ent, flexible workflow to process raw sequencing reads into a high-quality expression dataset ready for downstream analysis. scater provides a rich suite of plotting tools for single-cell data and a flex-ible data structure that is compatible with existing tools and can be used as infrastructure for future software development. Availability and Implementation: The open-source code, along with installation instructions, vi-gnettes and case studies, is available through Bioconductor at},
author = {McCarthy, Davis J. and Campbell, Kieran R. and Lun, Aaron T.L. and Wills, Quin F.},
doi = {10.1093/bioinformatics/btw777},
file = {:Users/nils/Documents/Mendeley Desktop/btw777.pdf:pdf},
isbn = {1367-4811 (Electronic)
1367-4803 (Linking)},
issn = {14602059},
journal = {Bioinformatics},
number = {8},
pages = {1179--1186},
pmid = {28088763},
title = {{Scater: Pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R}},
volume = {33},
year = {2017}
}
@article{Katayama2013,
abstract = {MOTIVATION: Recent transcriptome studies have revealed that total transcript numbers vary by cell type and condition; therefore, the statistical assumptions for single-cell transcriptome studies must be revisited. SAMstrt is an extension code for SAMseq, which is a statistical method for differential expression, to enable spike-in normalization and statistical testing based on the estimated absolute number of transcripts per cell for single-cell RNA-seq methods. Availability and Implementation: SAMstrt is implemented on R and available in github (https://github.com/shka/R-SAMstrt).$\backslash$n$\backslash$nCONTACT: [email protected]},
author = {Katayama, Shintaro and T{\"{o}}h{\"{o}}nen, Virpi and Linnarsson, Sten and Kere, Juha},
doi = {10.1093/bioinformatics/btt511},
file = {:Users/nils/Documents/Mendeley Desktop/Katayama et al. - 2013 - SAMstrt Statistical test for differential expression in single-cell transcriptome with spike-in normalization.pdf:pdf},
isbn = {1367-4811 (Electronic)
1367-4803 (Linking)},
issn = {13674803},
journal = {Bioinformatics},
number = {22},
pages = {2943--2945},
pmid = {23995393},
title = {{SAMstrt: Statistical test for differential expression in single-cell transcriptome with spike-in normalization}},
volume = {29},
year = {2013}
}
@article{Robinson2010,
title = {{{edgeR}}: A {{Bioconductor}} Package for Differential Expression Analysis of Digital Gene Expression Data},
shorttitle = {{{edgeR}}},
author = {Robinson, M. D. and McCarthy, D. J. and Smyth, G. K.},
year = {2010},
month = jan,
journal = {Bioinformatics},
volume = {26},
number = {1},
pages = {139--140},
issn = {1367-4803, 1460-2059},
doi = {10.1093/bioinformatics/btp616},
abstract = {Summary: It is expected that emerging digital gene expression (DGE) technologies will overtake microarray technologies in the near future for many functional genomics applications. One of the fundamental data analysis tasks, especially for gene expression studies, involves determining whether there is evidence that counts for a transcript or exon are significantly different across experimental conditions. edgeR is a Bioconductor software package for examining differential expression of replicated count data. An overdispersed Poisson model is used to account for both biological and technical variability. Empirical Bayes methods are used to moderate the degree of overdispersion across transcripts, improving the reliability of inference. The methodology can be used even with the most minimal levels of replication, provided at least one phenotype or experimental condition is replicated. The software may have other applications beyond sequencing data, such as proteome peptide count data.},
langid = {english},
file = {/home/alan/Documents/Papers/Robinson et al. - 2010 - edgeR a Bioconductor package for differential exp.pdf}
}
@article{Carpenter2017,
title={Stan: A Probabilistic Programming Language},
volume={76},
url={https://www.jstatsoft.org/index.php/jss/article/view/v076i01},
doi={10.18637/jss.v076.i01},
abstract={Stan is a probabilistic programming language for specifying statistical models. A Stan program imperatively defines a log probability function over parameters conditioned on specified data and constants. As of version 2.14.0, Stan provides full Bayesian inference for continuous-variable models through Markov chain Monte Carlo methods such as the No-U-Turn sampler, an adaptive form of Hamiltonian Monte Carlo sampling. Penalized maximum likelihood estimates are calculated using optimization methods such as the limited memory Broyden-Fletcher-Goldfarb-Shanno algorithm. Stan is also a platform for computing log densities and their gradients and Hessians, which can be used in alternative algorithms such as variational Bayes, expectation propagation, and marginal inference using approximate integration. To this end, Stan is set up so that the densities, gradients, and Hessians, along with intermediate quantities of the algorithm such as acceptance probabilities, are easily accessible. Stan can be called from the command line using the cmdstan package, through R using the rstan package, and through Python using the pystan package. All three interfaces support sampling and optimization-based inference with diagnostics and posterior analysis. rstan and pystan also provide access to log probabilities, gradients, Hessians, parameter transforms, and specialized plotting.},
number={1},
journal={Journal of Statistical Software},
author={Carpenter, Bob and Gelman, Andrew and Hoffman, Matthew D. and Lee, Daniel and Goodrich, Ben and Betancourt, Michael and Brubaker, Marcus and Guo, Jiqiang and Li, Peter and Riddell, Allen},
year={2017},
pages={1--32}
}
@article{Bacher2016,
title = {Design and Computational Analysis of Single-Cell {{RNA-sequencing}} Experiments},
author = {Bacher, Rhonda and Kendziorski, Christina},
year = {2016},
month = dec,
journal = {Genome Biology},
volume = {17},
number = {1},
pages = {63},
issn = {1474-760X},
doi = {10.1186/s13059-016-0927-y},
urldate = {2023-08-05},
langid = {english},
file = {/home/alan/Zotero/storage/M4EQEK5E/Bacher and Kendziorski - 2016 - Design and computational analysis of single-cell R.pdf}
}
@article{Delmans2016,
abstract = {BACKGROUND The advent of high throughput RNA-seq at the single-cell level has opened up new opportunities to elucidate the heterogeneity of gene expression. One of the most widespread applications of RNA-seq is to identify genes which are differentially expressed between two experimental conditions. RESULTS We present a discrete, distributional method for differential gene expression (D(3)E), a novel algorithm specifically designed for single-cell RNA-seq data. We use synthetic data to evaluate D(3)E, demonstrating that it can detect changes in expression, even when the mean level remains unchanged. Since D(3)E is based on an analytically tractable stochastic model, it provides additional biological insights by quantifying biologically meaningful properties, such as the average burst size and frequency. We use D(3)E to investigate experimental data, and with the help of the underlying model, we directly test hypotheses about the driving mechanism behind changes in gene expression. CONCLUSION Evaluation using synthetic data shows that D(3)E performs better than other methods for identifying differentially expressed genes since it is designed to take full advantage of the information available from single-cell RNA-seq experiments. Moreover, the analytical model underlying D(3)E makes it possible to gain additional biological insights.},
author = {Delmans, Mihails and Hemberg, Martin},
doi = {10.1186/s12859-016-0944-6},
issn = {1471-2105},
journal = {BMC bioinformatics},
keywords = {110,1186,17,2016,bioinformatics,doi 10,mans and hemberg bmc,s12859-016-0944-6},
number = {1},
pages = {110},
pmid = {26927822},
publisher = {BMC Bioinformatics},
title = {{Discrete distributional differential expression (D(3)E) - a tool for gene expression analysis of single-cell RNA-seq data.}},
url = {http://dx.doi.org/10.1186/s12859-016-0944-6{\%}5Cnhttp://www.ncbi.nlm.nih.gov/pubmed/26927822{\%}5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC4772470},
volume = {17},
year = {2016}
}
@article{Lun2016,
author = {Lun, Aaron T. L. and McCarthy, Davis J. and Marioni, John C.},
doi = {10.12688/f1000research.9501.1},
file = {:Users/nils/Documents/Mendeley Desktop/Lun, McCarthy, Marioni - 2016 - A step-by-step workflow for basic analyses of single-cell RNA-seq data.pdf:pdf},
issn = {2046-1402},
journal = {F1000Research},
number = {2122},
title = {{A step-by-step workflow for basic analyses of single-cell RNA-seq data}},
volume = {5},
year = {2016}
}
@article{Vallejos2015,
author = {Vallejos, Catalina A. and Marioni, John C. and Richardson, Sylvia},
doi = {10.1371/journal.pcbi.1004333},
file = {:Users/nils/Documents/Mendeley Desktop/Vallejos, Marioni, Richardson - 2015 - BASiCS Bayesian Analysis of Single-Cell Sequencing Data.pdf:pdf},
issn = {1553-7358},
journal = {PLOS Computational Biology},
pages = {e1004333},
title = {{BASiCS: Bayesian analysis of single-cell sequencing data}},
url = {http://dx.plos.org/10.1371/journal.pcbi.1004333},
volume = {11},
year = {2015}
}
@misc{Brennecke2013,
abstract = {Single-cell RNA-seq can yield valuable insights about the variability within a population of seemingly homogeneous cells. We developed a quantitative statistical method to distinguish true biological variability from the high levels of technical noise in single-cell experiments. Our approach quantifies the statistical significance of observed cell-to-cell variability in expression strength on a gene-by-gene basis. We validate our approach using two independent data sets from Arabidopsis thaliana and Mus musculus.},
author = {Brennecke, Philip and Anders, Simon and Kim, Jong Kyoung and Ko{\l}odziejczyk, Aleksandra A and Zhang, Xiuwei and Proserpio, Valentina and Baying, Bianka and Benes, Vladimir and Teichmann, Sarah a and Marioni, John C and Heisler, Marcus G},
booktitle = {Nature methods},
doi = {10.1038/nmeth.2645},
file = {:Users/nils/Documents/Mendeley Desktop/Brennecke et al. - 2013 - Accounting for technical noise in single-cell RNA-seq experiments(4).pdf:pdf},
isbn = {0000000000},
issn = {1548-7105},
keywords = {RNA,RNA: methods,Sequence Analysis,Single-Cell Analysis},
number = {11},
pages = {1093--5},
pmid = {24056876},
title = {{Accounting for technical noise in single-cell RNA-seq experiments.}},
url = {http://www.ncbi.nlm.nih.gov/pubmed/24056876},
volume = {10},
year = {2013}
}
@article{Lonnberg2017,
abstract = {Differentiation of na{\"{i}}ve CD4(+) T cells into functionally distinct T helper subsets is crucial for the orchestration of immune responses. Due to extensive heterogeneity and multiple overlapping transcriptional programs in differentiating T cell populations, this process has remained a challenge for systematic dissection in vivo. By using single-cell transcriptomics and computational analysis using a temporal mixtures of Gaussian processes model, termed GPfates, we reconstructed the developmental trajectories of Th1 and Tfh cells during blood-stage Plasmodium infection in mice. By tracking clonality using endogenous TCR sequences, we first demonstrated that Th1/Tfh bifurcation had occurred at both population and single-clone levels. Next, we identified genes whose expression was associated with Th1 or Tfh fates, and demonstrated a T-cell intrinsic role for Galectin-1 in supporting a Th1 differentiation. We also revealed the close molecular relationship between Th1 and IL-10-producing Tr1 cells in this infection. Th1 and Tfh fates emerged from a highly proliferative precursor that upregulated aerobic glycolysis and accelerated cell cycling as cytokine expression began. Dynamic gene expression of chemokine receptors around bifurcation predicted roles for cell-cell in driving Th1/Tfh fates. In particular, we found that precursor Th cells were coached towards a Th1 but not a Tfh fate by inflammatory monocytes. Thus, by integrating genomic and computational approaches, our study has provided two unique resources, a database www.PlasmoTH.org, which facilitates discovery of novel factors controlling Th1/Tfh fate commitment, and more generally, GPfates, a modelling framework for characterizing cell differentiation towards multiple fates.},
author = {L{\"{o}}nnberg, Tapio and Svensson, Valentine and James, Kylie R. and Fernandez-Ruiz, Daniel and Sebina, Ismail and Montandon, Ruddy and Soon, Megan S. F. and Fogg, Lily G. and Nair, Arya Sheela and Liligeto, Urijah N. and Stubbington, Michael J. T. and Ly, Lam-Ha and Bagger, Frederik Otzen and Zwiessele, Max and Lawrence, Neil D. and Souza-Fonseca-Guimaraes, Fernando and Bunn, Patrick T. and Engwerda, Christian R. and Heath, William R. and Billker, Oliver and Stegle, Oliver and Haque, Ashraful and Teichmann, Sarah A.},
doi = {10.1126/sciimmunol.aal2192},
file = {:Users/nils/Documents/Mendeley Desktop/lnnberg2017.pdf:pdf},
issn = {2470-9468},
journal = {Science Immunology},
number = {9},
pages = {eaal2192},
pmid = {28345074},
title = {{Single-cell RNA-seq and computational analysis using temporal mixture modeling resolves Th1/Tfh fate bifurcation in malaria}},
url = {http://immunology.sciencemag.org/lookup/doi/10.1126/sciimmunol.aal2192},
volume = {2},
year = {2017}
}
@article{Rna2005,
abstract = {The External RNA Control Consortium (ERCC) is an ad-hoc group with approximately 70 members from private, public, and academic organizations. The group is developing a set of external RNA control transcripts that can be used to assess technical performance in gene expression assays. The ERCC is now initiating the Testing Phase of the project, during which candidate external RNA controls will be evaluated in both microarray and QRT-PCR gene expression platforms. This document describes the proposed experiments and informatics process that will be followed to test and qualify individual controls. The ERCC is distributing this description of the proposed testing process in an effort to gain consensus and to encourage feedback from the scientific community. On October 4-5, 2005, the ERCC met to further review the document, clarify ambiguities, and plan next steps. A summary of this meeting and changes to the test plan are provided as an appendix to this manuscript.},
author = {{External RNA Controls Consortium}},
doi = {10.1186/1471-2164-6-150},
file = {:Users/nils/Documents/Mendeley Desktop/External RNA Controls Consortium - 2005 - Proposed methods for testing and selecting the ERCC external RNA controls.pdf:pdf},
isbn = {1471-2164 (Electronic) 1471-2164 (Linking)},
issn = {1471-2164},
journal = {BMC Genomics},
number = {June 2004},
pages = {150},
pmid = {16266432},
title = {{Proposed methods for testing and selecting the ERCC external RNA controls.}},
volume = {6},
year = {2005}
}
@article{Tung2016,
abstract = {Single cell RNA sequencing (scRNA-seq) can be used to characterize variation in gene expression levels at high resolution. However, the sources of experimental noise in scRNA-seq are not yet well understood. We investigated the technical variation associated with sample processing using the single cell Fluidigm C1 platform. To do so, we processed three C1 replicates from three human induced pluripotent stem cell (iPSC) lines. We added unique molecular identifiers (UMIs) to all samples, to account for amplification bias. We found that the major source of variation in the gene expression data was driven by genotype, but we also observed substantial variation between the technical replicates. We observed that the conversion of reads to molecules using the UMIs was impacted by both biological and technical variation, indicating that UMI counts are not an unbiased estimator of gene expression levels. Based on our results, we suggest a framework for effective scRNA-seq studies.},
author = {Tung, Po-Yuan and Blischak, John D and Hsiao, Chiaowen and Knowles, David A and Burnett, Jonathan E and Pritchard, Jonathan K and Gilad, Yoav},
doi = {10.1101/062919},
file = {:Users/nils/Documents/Mendeley Desktop/062919.full-2.pdf:pdf},
journal = {bioRxiv},
pages = {062919},
title = {{Batch effects and the effective design of single-cell gene expression studies}},
url = {http://biorxiv.org/lookup/doi/10.1101/062919},
year = {2016}
}
@article{Grun2014,
abstract = {Single-cell transcriptomics has recently emerged as a powerful technology to explore gene expression heterogeneity among single cells. Here we identify two major sources of technical variability: sampling noise and global cell-to-cell variation in sequencing efficiency. We propose noise models to correct for this, which we validate using single-molecule FISH. We demonstrate that gene expression variability in mouse embryonic stem cells depends on the culture condition.},
author = {Gr{\"{u}}n, Dominic and Kester, Lennart and van Oudenaarden, Alexander},
doi = {10.1038/nmeth.2930},
file = {:Users/nils/Documents/Mendeley Desktop/Gr{\"{u}}n, Kester, van Oudenaarden - 2014 - Validation of noise models for single-cell transcriptomics.pdf:pdf},
issn = {1548-7105},
journal = {Nature methods},
keywords = {Animals,Embryonic Stem Cells,Embryonic Stem Cells: metabolism,Gene Expression Profiling,Gene Expression Profiling: methods,Gene Expression Profiling: standards,Gene Expression Regulation,Mice,Models, Biological,Observer Variation,Selection Bias,Sequence Analysis, RNA,Signal-To-Noise Ratio,Transcription Factors,Transcription Factors: metabolism},
month = {jun},
number = {6},
pages = {637--40},
pmid = {24747814},
title = {{Validation of noise models for single-cell transcriptomics.}},
url = {http://www.ncbi.nlm.nih.gov/pubmed/24747814},
volume = {11},
year = {2014}
}
@article{Proserpio2016,
author = {Proserpio, Valentina and Piccolo, Andrea and Haim-Vilmovsky, Liora and Kar, Gozde and L{\"{o}}nnberg, Tapio and Svensson, Valentine and Pramanik, Jhuma and Natarajan, Kedar Nath and Zhai, Weichao and Zhang, Xiuwei and Donati, Giacomo and Kayikci, Melis and Kotar, Jurij and McKenzie, Andrew N. J. and Montandon, Ruddy and Billker, Oliver and Woodhouse, Steven and Cicuta, Pietro and Nicodemi, Mario and Teichmann, Sarah A.},
doi = {10.1186/s13059-016-0957-5},
file = {:Users/nils/Documents/Mendeley Desktop/Proserpio et al. - 2016 - Single-cell analysis of CD4 T-cell differentiation reveals three major cell states and progressive acceleratio.pdf:pdf},
isbn = {10.1186/s13059-016-0957-5},
issn = {1474-760X},
journal = {Genome Biology},
keywords = {CD4+ T cells,Adaptive immunity,Single-cell RNA-seq,adaptive immunity,cd4,cell cycle,differentiation,live imaging,single-cell rna-seq,t cells},
number = {1},
pages = {103},
pmid = {27176874},
publisher = {Genome Biology},
title = {{Single-cell analysis of CD4+ T-cell differentiation reveals three major cell states and progressive acceleration of proliferation}},
url = {http://genomebiology.biomedcentral.com/articles/10.1186/s13059-016-0957-5},
volume = {17},
year = {2016}
}
@article{Kharchenko2014,
abstract = {Single-cell data provide a means to dissect the composition of complex tissues and specialized cellular environments. However, the analysis of such measurements is complicated by high levels of technical noise and intrinsic biological variability. We describe a probabilistic model of expression-magnitude distortions typical of single-cell RNA-sequencing measurements, which enables detection of differential expression signatures and identification of subpopulations of cells in a way that is more tolerant of noise.},
author = {Kharchenko, Peter V and Silberstein, Lev and Scadden, David T},
doi = {10.1038/nmeth.2967},
file = {:Users/nils/Documents/Mendeley Desktop/Kharchenko, Silberstein, Scadden - 2014 - Bayesian approach to single-cell differential expression analysis.pdf:pdf},
issn = {1548-7105},
journal = {Nature methods},
month = {jul},
number = {7},
pages = {740--2},
pmid = {24836921},
title = {{Bayesian approach to single-cell differential expression analysis.}},
url = {http://www.ncbi.nlm.nih.gov/pubmed/24836921},
volume = {11},
year = {2014}
}
@article{Eling2017,
abstract = {Cell-to-cell transcriptional variability in otherwise homogeneous cell populations plays a crucial role in tissue function and development. Single-cell RNA sequencing can characterise this variability in a transcriptome-wide manner. However, technical variation and the confounding between variability and mean expression estimates hinders meaningful comparison of expression variability between cell populations. To address this problem, we introduce a novel analysis approach that extends the BASiCS statistical framework to derive a residual measure of variability that is not confounded by mean expression. Moreover, we introduce a new and robust procedure for quantifying technical noise in experiments where technical spike-in molecules are not available. We illustrate how our method provides biological insight into the dynamics of cell-to-cell expression variability, highlighting a synchronisation of the translational machinery in immune cells upon activation. Additionally, our approach identifies new patterns of variability across CD4+ T cell differentiation.},
author = {Eling, Nils and Richard, Arianne C. and Richardson, Sylvia and Marioni, John C. and Vallejos, Catalina A.},
doi = {10.1101/237214},
file = {:Users/nils/Documents/Mendeley Desktop/237214.full.pdf:pdf},
journal = {bioRxiv},
pages = {237214},
title = {{Robust expression variability testing reveals heterogeneous T cell responses}},
url = {https://www.biorxiv.org/content/early/2017/12/21/237214.full.pdf+html},
year = {2017}
}
@article{Goolam2016,
abstract = {The major and essential objective of pre-implantation development is to establish embryonic and extra-embryonic cell fates. To address when and how this fundamental process is initiated in mammals, we characterize transcriptomes of all individual cells throughout mouse pre-implantation development. This identifies targets of master pluripotency regulators Oct4 and Sox2 as being highly heterogeneously expressed between blastomeres of the 4-cell embryo, with Sox21 showing one of the most heterogeneous expression profiles. Live-cell tracking demonstrates that cells with decreased Sox21 yield more extra-embryonic than pluripotent progeny. Consistently, decreasing Sox21 results in premature upregulation of the differentiation regulator Cdx2, suggesting that Sox21 helps safeguard pluripotency. Furthermore, Sox21 is elevated following increased expression of the histone H3R26-methylase CARM1 and is lowered following CARM1 inhibition, indicating the importance of epigenetic regulation. Therefore, our results indicate that heterogeneous gene expression, as early as the 4-cell stage, initiates cell-fate decisions by modulating the balance of pluripotency and differentiation.},
archivePrefix = {arXiv},
arxivId = {cs/9605103},
author = {Goolam, Mubeen and Scialdone, Antonio and Graham, Sarah J L and MacAulay, Iain C. and Jedrusik, Agnieszka and Hupalowska, Anna and Voet, Thierry and Marioni, John C. and Zernicka-Goetz, Magdalena},
doi = {10.1016/j.cell.2016.01.047},
eprint = {9605103},
file = {:Users/nils/Documents/Mendeley Desktop/main.pdf:pdf},
isbn = {0-7803-3213-X},
issn = {10974172},
journal = {Cell},
number = {1},
pages = {61--74},
pmid = {27015307},
primaryClass = {cs},
publisher = {The Authors},
title = {{Heterogeneity in Oct4 and Sox2 Targets Biases Cell Fate in 4-Cell Mouse Embryos}},
url = {http://dx.doi.org/10.1016/j.cell.2016.01.047},
volume = {165},
year = {2016}
}
@article{Ibarra-Soria2018a,
abstract = {During gastrulation, cell types from all three germ layers are specified and the basic body plan is established
1
. However, molecular analysis of this key developmental stage has been hampered by limited cell numbers and a paucity of markers. Single-cell RNA sequencing circumvents these problems, but has so far been limited to specific organ systems
2
. Here, we report single-cell transcriptomic characterization of {\textgreater}20,000 cells immediately following gastrulation at E8.25 of mouse development. We identify 20 major cell types, which frequently contain substructure, including three distinct signatures in early foregut cells. Pseudo-space ordering of somitic progenitor cells identifies dynamic waves of transcription and candidate regulators, which are validated by molecular characterization of spatially resolved regions of the embryo. Within the endothelial population, cells that transition from haemogenic endothelial to erythro-myeloid progenitors specifically express Alox5 and its co-factor Alox5ap, which control leukotriene production. Functional assays using mouse embryonic stem cells demonstrate that leukotrienes promote haematopoietic progenitor cell generation. Thus, this comprehensive single-cell map can be exploited to reveal previously unrecognized pathways that contribute to tissue development.},
author = {Ibarra-Soria, Ximena and Jawaid, Wajid and Pijuan-Sala, Blanca and Ladopoulos, Vasileios and Scialdone, Antonio and J{\"{o}}rg, David J. and Tyser, Richard C.V. and Calero-Nieto, Fernando J. and Mulas, Carla and Nichols, Jennifer and Vallier, Ludovic and Srinivas, Shankar and Simons, Benjamin D. and G{\"{o}}ttgens, Berthold and Marioni, John C.},
doi = {10.1038/s41556-017-0013-z},
file = {:Users/nils/Documents/Mendeley Desktop/ibarrasoria2018.pdf:pdf},
issn = {14764679},
journal = {Nature Cell Biology},
number = {2},
pages = {127--134},
pmid = {29311656},
title = {{Defining murine organogenesis at single-cell resolution reveals a role for the leukotriene pathway in regulating blood progenitor formation}},
volume = {20},
year = {2018}
}
@article{Kolodziejczyk2015a,
author = {Kolodziejczyk, Aleksandra A. and Kim, Jong Kyoung and Tsang, Jason C.H. and Ilicic, Tomislav and Henriksson, Johan and Natarajan, Kedar N. and Tuck, Alex C. and Gao, Xuefei and B{\"{u}}hler, Marc and Liu, Pentao and Marioni, John C. and Teichmann, Sarah A.},
doi = {10.1016/j.stem.2015.09.011},
file = {:Users/nils/Documents/Mendeley Desktop/Kolodziejczyk et al. - 2015 - Single cell RNA-sequencing of pluripotent states unlocks modular transcriptional variation.pdf:pdf},
issn = {19345909},
journal = {Cell Stem Cell},
pages = {471--485},
title = {{Single cell RNA-sequencing of pluripotent states unlocks modular transcriptional variation}},
url = {http://linkinghub.elsevier.com/retrieve/pii/S193459091500418X},
volume = {17},
year = {2015}
}
@article{Stegle2015,
author = {Stegle, Oliver and Teichmann, Sarah a. and Marioni, John C.},
doi = {10.1038/nrg3833},
file = {:Users/nils/Documents/Mendeley Desktop/Stegle, Teichmann, Marioni - 2015 - Computational and analytical challenges in single-cell transcriptomics.pdf:pdf},
issn = {1471-0056},
journal = {Nature Reviews Genetics},
month = {jan},
number = {3},
pages = {133--145},
publisher = {Nature Publishing Group},
title = {{Computational and analytical challenges in single-cell transcriptomics}},
url = {http://www.nature.com/doifinder/10.1038/nrg3833},
volume = {16},
year = {2015}
}
@article{Anders2010,
author = {Anders, Simon and Huber, Wolfgang},
doi = {10.1186/gb-2010-11-10-r106},
file = {:Users/nils/Documents/Mendeley Desktop/Anders, Huber - 2010 - Differential expression analysis for sequence count data.pdf:pdf},
issn = {1465-6906},
journal = {Genome Biology},
number = {10},
pages = {R106},
publisher = {BioMed Central Ltd},
title = {{Differential expression analysis for sequence count data}},
url = {http://genomebiology.com/2010/11/10/R106},
volume = {11},
year = {2010}
}
@article{Eberwine2015,
abstract = {The advent of single cell transcriptome analysis has permitted the discovery of cell-to-cell variation in transcriptome expression of even presumptively identical cells. We hypothesize that this variability reflects a many-to-one relation between transcriptome states and the phenotype of a cell. In this relation, the molecular ratios of the subsets of RNA are determined by the stoichiometric constraints of the cell systems, which underdetermine the transcriptome state. Furthermore, the variability is, in part, induced by the tissue context and is important for system-level function. This theory is analogous to theories of literary deconstruction, where multiple 'signifiers' work in opposition to one another to create meaning. By analogy, transcriptome phenotypes should be defined as subsets of RNAs comprising selected RNA systems where the system-associated RNAs are balanced with each other to produce the associated cellular function. This idea provides a framework for understanding cellular heterogeneity in phenotypic responses to variant conditions, such as disease challenge. Comparisons of transcriptomes between seemingly identical cells show that the transcriptomes have significant biological variation.Cells exhibit 'many-to-one' sets of equally functional transcriptional states that are defined, in part, by pathway constraints that are underspecified in comparison to the number of genes.Cellular contextualization occurs from the influence of the microenvironment and local interactions, suggesting that variation itself is important for system-level function.},
author = {Eberwine, James and Kim, Junhyong},
doi = {10.1016/j.tcb.2015.07.004},
file = {:Users/nils/Documents/Mendeley Desktop/Eberwine, Kim - 2015 - Cellular Deconstruction Finding Meaning in Individual Cell Variation.pdf:pdf},
issn = {18793088},
journal = {Trends in Cell Biology},
keywords = {Biological constraints,Phenotype,Transcriptome},
number = {10},
pages = {569--578},
pmid = {26410403},
publisher = {Elsevier Ltd},
title = {{Cellular Deconstruction: Finding Meaning in Individual Cell Variation}},
url = {http://dx.doi.org/10.1016/j.tcb.2015.07.004},
volume = {25},
year = {2015}
}
@book{Gelman2014,
author = {Gelman, Andrew and Carlin, John and Stern, Hal and Dunson, David B and Vehtari, Aki and Rubin, Donald B},
isbn = {978-1439840955},
pages = {286--288},
publisher = {CRC Press},
title = {{Bayesian Data Analysis}},
year = {2014}
}
@article{CowlesCarlin1996,
author = { Mary Kathryn Cowles and Bradley P. Carlin },
title = {Markov Chain Monte Carlo Convergence Diagnostics: A Comparative Review},
journal = {Journal of the American Statistical Association},
volume = {91},
number = {434},
pages = {883-904},
year = {1996},
publisher = {Taylor & Francis},
doi = {10.1080/01621459.1996.10476956},
URL = { https://amstat.tandfonline.com/doi/abs/10.1080/01621459.1996.10476956},
eprint = { https://amstat.tandfonline.com/doi/pdf/10.1080/01621459.1996.10476956}
}
@article{BrooksGelman1998,
author = { Stephen P. Brooks and Andrew Gelman },
title = {General Methods for Monitoring Convergence of Iterative Simulations},
journal = {Journal of Computational and Graphical Statistics},
volume = {7},
number = {4},
pages = {434-455},
year = {1998},
publisher = {Taylor & Francis},
doi = {10.1080/10618600.1998.10474787},
URL = { https://amstat.tandfonline.com/doi/abs/10.1080/10618600.1998.10474787},
eprint = { https://amstat.tandfonline.com/doi/pdf/10.1080/10618600.1998.10474787}
}
@article{CODA2006,
volume = {6},
number = {1},
month = {March},
author = {Martyn Plummer and Nicky Best and Kate Cowles and Karen Vines},
title = {CODA: convergence diagnosis and output analysis for MCMC},
journal = {R News},
pages = {7--11},
year = {2006},
url = {http://oro.open.ac.uk/22547/},
}
@article{Prakadan2017,
abstract = {Recent advances in cellular profiling have demonstrated substantial heterogeneity in the behaviour of cells once deemed 'identical', challenging fundamental notions of cell 'type' and 'state'. Not surprisingly, these findings have elicited substantial interest in deeply characterizing the diversity, interrelationships and plasticity among cellular phenotypes. To explore these questions, experimental platforms are needed that can extensively and controllably profile many individual cells. Here, microfluidic structures - whether valve-, droplet- or nanowell-based - have an important role because they can facilitate easy capture and processing of single cells and their components, reducing labour and costs relative to conventional plate-based methods while also improving consistency. In this article, we review the current state-of-the-art methodologies with respect to microfluidics for mammalian single-cell 'omics' and discuss challenges and future opportunities.},
archivePrefix = {arXiv},
arxivId = {10.1038/nrg.2017.15},
author = {Prakadan, Sanjay M. and Shalek, Alex K. and Weitz, David A.},
doi = {10.1038/nrg.2017.15},
eprint = {nrg.2017.15},
file = {:Users/nils/Documents/Mendeley Desktop/Prakadan, Shalek, Weitz - 2017 - Scaling by shrinking empowering single-cell 'omics' with microfluidic devices.pdf:pdf},
isbn = {1471-0056},
issn = {1471-0056},
journal = {Nature Reviews Genetics},
number = {6},
pages = {345--361},
pmid = {28392571},
primaryClass = {10.1038},
title = {{Scaling by shrinking: empowering single-cell 'omics' with microfluidic devices}},
url = {http://www.nature.com/doifinder/10.1038/nrg.2017.15},
volume = {18},
year = {2017}
}
@article{Patange2018,
abstract = {Gene expression varies across cells in a population or a tissue. This heterogeneity has come into sharp focus in recent years through developments in new imaging and sequencing technologies. However, our ability to measure variation has outpaced our ability to interpret it. Much of the variability may arise from random effects occurring in the processes of gene expression (transcription, RNA processing and decay, translation). The molecular basis of these effects is largely unknown. Likewise, a functional role of this variability in growth, differentiation and disease has only been elucidated in a few cases. In this review, we highlight recent experimental and theoretical advances for measuring and analyzing stochastic variation.},
author = {Patange, Simona and Girvan, Michelle and Larson, Daniel R.},
doi = {10.1016/j.coisb.2017.11.011},
file = {:Users/nils/Documents/Mendeley Desktop/Patange, Girvan, Larson - 2018 - Single-cell systems biology Probing the basic unit of information flow.pdf:pdf},
issn = {24523100},
journal = {Current Opinion in Systems Biology},
keywords = {Live cell imaging,Single-cell gene expression,Single-molecule RNA FISH,scRNA-seq},
pages = {7--15},
publisher = {Elsevier Ltd},
title = {{Single-cell systems biology: Probing the basic unit of information flow}},
url = {https://doi.org/10.1016/j.coisb.2017.11.011},
volume = {8},
year = {2018}
}
@article{Villani2017,
abstract = {Dendritic cells (DCs) and monocytes play a central role in pathogen sensing, phagocytosis, and antigen presentation and consist of multiple specialized subtypes. However, their identities and interrelationships are not fully understood. Using unbiased single-cell RNA sequencing (RNA-seq) of {\~{}}2400 cells, we identified six human DCs and four monocyte subtypes in human blood. Our study reveals a new DC subset that shares properties with plasmacytoid DCs (pDCs) but potently activates T cells, thus redefining pDCs; a new subdivision within the CD1C(+) subset of DCs; the relationship between blastic plasmacytoid DC neoplasia cells and healthy DCs; and circulating progenitor of conventional DCs (cDCs). Our revised taxonomy will enable more accurate functional and developmental analyses as well as immune monitoring in health and disease.},
archivePrefix = {arXiv},
arxivId = {15334406},
author = {Villani, Alexandra Chlo{\'{e}} and Satija, Rahul and Reynolds, Gary and Sarkizova, Siranush and Shekhar, Karthik and Fletcher, James and Griesbeck, Morgane and Butler, Andrew and Zheng, Shiwei and Lazo, Suzan and Jardine, Laura and Dixon, David and Stephenson, Emily and Nilsson, Emil and Grundberg, Ida and McDonald, David and Filby, Andrew and Li, Weibo and {De Jager}, Philip L. and Rozenblatt-Rosen, Orit and Lane, Andrew A. and Haniffa, Muzlifah and Regev, Aviv and Hacohen, Nir},
doi = {10.1126/science.aah4573},
eprint = {15334406},
file = {:Users/nils/Documents/Mendeley Desktop/Villani et al. - 2017 - Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors.pdf:pdf},
isbn = {1095-9203 (Electronic) 0036-8075 (Linking)},
issn = {10959203},
journal = {Science},
number = {6335},
pmid = {28428369},
title = {{Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors}},
volume = {356},
year = {2017}
}
@article{Zheng2017,
abstract = {Characterizing the transcriptome of individual cells is fundamental to understanding complex biological systems. We describe a droplet-based system that enables 3′ mRNA counting of up to tens of thousands of single cells per sample. Cell encapsulation in droplets takes place in {\~{}}6 minutes, with {\~{}}50{\%} cell capture efficiency, up to 8 samples at a time. The speed and efficiency allow the processing of precious samples while minimizing stress to cells. To demonstrate the system′s technical performance and its applications, we collected transcriptome data from {\~{}}¼ million single cells across 29 samples. First, we validate the sensitivity of the system and its ability to detect rare populations using cell lines and synthetic RNAs. Then, we profile 68k peripheral blood mononuclear cells (PBMCs) to demonstrate the system′s ability to characterize large immune populations. Finally, we use sequence variation in the transcriptome data to determine host and donor chimerism at single cell resolution in bone marrow mononuclear cells (BMMCs) of transplant patients. This analysis enables characterization of the complex interplay between donor and host cells and monitoring of treatment response. This high-throughput system is robust and enables characterization of diverse biological systems with single cell mRNA analysis.},
archivePrefix = {arXiv},
arxivId = {065912},
author = {Zheng, Grace X.Y. and Terry, Jessica M. and Belgrader, Phillip and Ryvkin, Paul and Bent, Zachary W. and Wilson, Ryan and Ziraldo, Solongo B. and Wheeler, Tobias D. and McDermott, Geoff P. and Zhu, Junjie and Gregory, Mark T. and Shuga, Joe and Montesclaros, Luz and Underwood, Jason G. and Masquelier, Donald A. and Nishimura, Stefanie Y. and Schnall-Levin, Michael and Wyatt, Paul W. and Hindson, Christopher M. and Bharadwaj, Rajiv and Wong, Alexander and Ness, Kevin D. and Beppu, Lan W. and Deeg, H. Joachim and McFarland, Christopher and Loeb, Keith R. and Valente, William J. and Ericson, Nolan G. and Stevens, Emily A. and Radich, Jerald P. and Mikkelsen, Tarjei S. and Hindson, Benjamin J. and Bielas, Jason H.},
doi = {10.1038/ncomms14049},
eprint = {065912},
file = {:Users/nils/Documents/Mendeley Desktop/ncomms14049.pdf:pdf},
isbn = {9780979806483},
issn = {20411723},
journal = {Nature Communications},
pages = {1--12},
pmid = {28091601},
publisher = {Nature Publishing Group},
title = {{Massively parallel digital transcriptional profiling of single cells}},
url = {http://dx.doi.org/10.1038/ncomms14049},
volume = {8},
year = {2017}
}
@article{Ibarra-Soria2018,
abstract = {During gastrulation, cell types from all three germ layers are specified and the basic body plan is established 1 . However, molecular analysis of this key developmental stage has been hampered by limited cell numbers and a paucity of markers. Single-cell RNA sequencing circumvents these problems, but has so far been limited to specific organ systems 2 . Here, we report single-cell transcriptomic characterization of {\textgreater}20,000 cells immediately following gastrulation at E8.25 of mouse development. We identify 20 major cell types, which frequently contain substructure, including three distinct signatures in early foregut cells. Pseudo-space ordering of somitic progenitor cells identifies dynamic waves of transcription and candidate regulators, which are validated by molecular characterization of spatially resolved regions of the embryo. Within the endothelial population, cells that transition from haemogenic endothelial to erythro-myeloid progenitors specifically express Alox5 and its co-factor Alox5ap, which control leukotriene production. Functional assays using mouse embryonic stem cells demonstrate that leukotrienes promote haematopoietic progenitor cell generation. Thus, this comprehensive single-cell map can be exploited to reveal previously unrecognized pathways that contribute to tissue development.},
author = {Ibarra-Soria, Ximena and Jawaid, Wajid and Pijuan-Sala, Blanca and Ladopoulos, Vasileios and Scialdone, Antonio and J{\"{o}}rg, David J. and Tyser, Richard C.V. and Calero-Nieto, Fernando J. and Mulas, Carla and Nichols, Jennifer and Vallier, Ludovic and Srinivas, Shankar and Simons, Benjamin D. and G{\"{o}}ttgens, Berthold and Marioni, John C.},
doi = {10.1038/s41556-017-0013-z},
file = {:Users/nils/Documents/Mendeley Desktop/ibarrasoria2018.pdf:pdf},
issn = {14764679},
journal = {Nature Cell Biology},
number = {2},
pages = {127--134},
pmid = {29311656},
title = {{Defining murine organogenesis at single-cell resolution reveals a role for the leukotriene pathway in regulating blood progenitor formation}},
volume = {20},
year = {2018}
}
@article{Wagner2018,
abstract = {As embryos develop, numerous cell types with distinct functions and morphologies arise from pluripotent cells. Three research groups have used single-cell RNA sequencing to analyze the transcriptional changes accompanying development of vertebrate embryos (see the Perspective by Harland). Wagner et al. sequenced the transcriptomes of more than 90,000 cells throughout zebrafish development to reveal how cells differentiate during axis patterning, germ layer formation, and early organogenesis. Farrell et al. profiled the transcriptomes of tens of thousands of embryonic cells and applied a computational approach to construct a branching tree describing the transcriptional trajectories that lead to 25 distinct zebrafish cell types. The branching tree revealed how cells change their gene expression as they become more and more specialized. Briggs et al. examined whole frog embryos, spanning zygotic genome activation through early organogenesis, to map cell states and differentiation across all cell lineages over time. These data and approaches pave the way for the comprehensive reconstruction of transcriptional trajectories during development. Science , this issue p. [981][1], p. [eaar3131][2], p. [eaar5780][3]; see also p. [967][4] [1]: /lookup/doi/10.1126/science.aar4362 [2]: /lookup/doi/10.1126/science.aar3131 [3]: /lookup/doi/10.1126/science.aar5780 [4]: /lookup/doi/10.1126/science.aat8413},
archivePrefix = {arXiv},
arxivId = {1101.5435},
author = {Wagner, Daniel E. and Weinreb, Caleb and Collins, Zach M. and Briggs, James A. and Megason, Sean G. and Klein, Allon M.},
doi = {10.1126/science.aar4362},
eprint = {1101.5435},
file = {:Users/nils/Documents/Mendeley Desktop/science.aar4362.full.pdf:pdf},
isbn = {9781605589077},
issn = {10959203},
journal = {Science},
pages = {1--12},
pmid = {29700229},
title = {{Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo}},
volume = {4362},
year = {2018}
}
@article{Pijuan-Sala2019,
abstract = {Across the animal kingdom, gastrulation represents a key developmental event during which embryonic pluripotent cells diversify into lineage-specific precursors that will generate the adult organism. Here we report the transcriptional profiles of 116,312 single cells from mouse embryos collected at nine sequential time points ranging from 6.5 to 8.5 days post-fertilization. We construct a molecular map of cellular differentiation from pluripotency towards all major embryonic lineages, and explore the complex events involved in the convergence of visceral and primitive streak-derived endoderm. Furthermore, we use single-cell profiling to show that Tal1−/− chimeric embryos display defects in early mesoderm diversification, and we thus demonstrate how combining temporal and transcriptional information can illuminate gene function. Together, this comprehensive delineation of mammalian cell differentiation trajectories in vivo represents a baseline for understanding the effects of gene mutations during development, as well as a roadmap for the optimization of in vitro differentiation protocols for regenerative medicine.},
author = {Pijuan-Sala, Blanca and Griffiths, Jonathan A. and Guibentif, Carolina and Hiscock, Tom W. and Jawaid, Wajid and Calero-Nieto, Fernando J. and Mulas, Carla and Ibarra-Soria, Ximena and Tyser, Richard C.V. and Ho, Debbie Lee Lian and Reik, Wolf and Srinivas, Shankar and Simons, Benjamin D. and Nichols, Jennifer and Marioni, John C. and G{\"{o}}ttgens, Berthold},
doi = {10.1038/s41586-019-0933-9},
file = {:Users/nils/Documents/Mendeley Desktop/[email protected]:pdf},
issn = {14764687},
journal = {Nature},
number = {7745},
pages = {490--495},
title = {{A single-cell molecular map of mouse gastrulation and early organogenesis}},
volume = {566},
year = {2019}
}
@article{Ohnishi2014,
abstract = {It is now recognized that extensive expression heterogeneities among cells precede the emergence of lineages in the early mammalian embryo. To establish a map of pluripotent epiblast (EPI) versus primitive endoderm (PrE) lineage segregation within the inner cell mass (ICM) of the mouse blastocyst, we characterized the gene expression profiles of individual ICM cells. Clustering analysis of the transcriptomes of 66 cells demonstrated that initially they are non-distinguishable. Early in the segregation, lineage-specific marker expression exhibited no apparent correlation, and a hierarchical relationship was established only in the late blastocyst. Fgf4 exhibited a bimodal expression at the earliest stage analysed, and in its absence, the differentiation of PrE and EPI was halted, indicating that Fgf4 drives, and is required for, ICM lineage segregation. These data lead us to propose a model where stochastic cell-to-cell expression heterogeneity followed by signal reinforcement underlies ICM lineage segregation by antagonistically separating equivalent cells.},
author = {Ohnishi, Yusuke and Huber, Wolfgang and Tsumura, Akiko and Kang, Minjung and Xenopoulos, Panagiotis and Kurimoto, Kazuki and Ole{\'{s}}, Andrzej K and Ara{\'{u}}zo-Bravo, Marcos J and Saitou, Mitinori and Hadjantonakis, Anna-Katerina and Hiiragi, Takashi},
doi = {10.1038/ncb2881},
file = {:Users/nils/Documents/Mendeley Desktop/Ohnishi et al. - 2014 - Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineag.pdf:pdf},
issn = {1476-4679},
journal = {Nature Cell Biology},
keywords = {Animals,Biological,Biological Markers,Biological Markers: metabolism,Blastocyst Inner Cell Mass,Blastocyst Inner Cell Mass: cytology,Blastocyst Inner Cell Mass: metabolism,Cell Lineage,Cell Lineage: drug effects,Cell Separation,Developmental,Endoderm,Endoderm: cytology,Endoderm: metabolism,Fibroblast Growth Factor 4,Fibroblast Growth Factor 4: metabolism,Gene Expression Profiling,Gene Expression Regulation,Germ Layers,Germ Layers: cytology,Germ Layers: metabolism,Mice,Models,Oligonucleotide Array Sequence Analysis,Polymerase Chain Reaction,Principal Component Analysis,Signal Transduction,Signal Transduction: genetics,Single-Cell Analysis},
number = {1},
pages = {27--37},
pmid = {24292013},
publisher = {Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
shorttitle = {Nat Cell Biol},
title = {{Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineages.}},
url = {http://dx.doi.org/10.1038/ncb2881},
volume = {16},
year = {2014}
}
@article{Kiselev2019,
abstract = {Single-cell RNA sequencing (scRNA-seq) allows researchers to collect large catalogues detailing the transcriptomes of individual cells. Unsupervised clustering is of central importance for the analysis of these data, as it is used to identify putative cell types. However, there are many challenges involved. We discuss why clustering is a challenging problem from a computational point of view and what aspects of the data make it challenging. We also consider the difficulties related to the biological interpretation and annotation of the identified clusters.},
author = {Kiselev, Vladimir Yu and Andrews, Tallulah S. and Hemberg, Martin},
doi = {10.1038/s41576-018-0088-9},
file = {:Users/nils/Documents/Mendeley Desktop/[email protected]:pdf},
issn = {1471-0064},
journal = {Nature Reviews Genetics 2018},
keywords = {Functional clustering,Gene expression analysis,Genome,RNA sequencing,Statistical methods,Transcriptomics,wide analysis of gene expression},
pmid = {30617341},
publisher = {Springer US},
title = {{Challenges in unsupervised clustering of single-cell RNA-seq data}},
url = {https://www.nature.com/articles/s41576-018-0088-9?utm{\_}source=feedburner{\&}utm{\_}medium=feed{\&}utm{\_}campaign=Feed{\%}3A+nrg{\%}2Frss{\%}2Fcurrent+{\%}28Nature+Reviews+Genetics+-+Issue{\%}29},
year = {2019}
}
@article{Elowitz2002,
abstract = {Clonal populations of cells exhibit substantial phenotypic variation. Such heterogeneity can be essential for many biological processes and is conjectured to arise from stochasticity, or noise, in gene expression. We constructed strains of Escherichia coli that enable detection of noise and discrimination between the two mechanisms by which it is generated. Both stochasticity inherent in the biochemical process of gene expression (intrinsic noise) and fluctuations in other cellular components (extrinsic noise) contribute substantially to overall variation. Transcription rate, regulatory dynamics, and genetic factors control the amplitude of noise. These results establish a quantitative foundation for modeling noise in genetic networks and reveal how low intracellular copy numbers of molecules can fundamentally limit the precision of gene regulation.},
author = {Elowitz, Michael B and Levine, Arnold J and Siggia, Eric D and Swain, Peter S},
doi = {10.1126/science.1070919},
file = {:Users/nils/Documents/Mendeley Desktop/science02.pdf:pdf},
isbn = {1095-9203 (Electronic)$\backslash$r0036-8075 (Linking)},
issn = {1095-9203},
journal = {Science},
keywords = {*Gene Expression Regulation,*Genes,Bacterial,Bacterial Proteins/biosynthesis/*genetics,Computer-Assisted,Culture Media,Escherichia coli/*genetics,Experimental noise,Fluorescence,Gene Dosage,Genes,Genetic,Image Processing,Isopropyl Thiogalactoside/metabolism/pharmacology,Luminescent Proteins/biosynthesis/*genetics,Noise in prokaryotes,Non-U.S. Gov't,P.H.S.,Plasmids,Promoter Regions (Genetics),Reporter,Repressor Proteins,Stochastic Processes,Support,Transcription,U.S. Gov't},
number = {5584},
pages = {1183--1186},
pmid = {12183631},
title = {{Stochastic gene expression in a single cell}},
url = {http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve{\&}db=PubMed{\&}dopt=Citation{\&}list{\_}uids=12183631},
volume = {297},
year = {2002}
}
@article{Zopf2013,
abstract = {The large variability in mRNA and protein levels found from both static and dynamic measurements in single cells has been largely attributed to random periods of transcription, often occurring in bursts. The cell cycle has a pronounced global role in affecting transcriptional and translational output, but how this influences transcriptional statistics from noisy promoters is unknown and generally ignored by current stochastic models. Here we show that variable transcription from the synthetic tetO promoter in S. cerevisiae is dominated by its dependence on the cell cycle. Real-time measurements of fluorescent protein at high expression levels indicate tetO promoters increase transcription rate ∼2-fold in S/G2/M similar to constitutive genes. At low expression levels, where tetO promoters are thought to generate infrequent bursts of transcription, we observe random pulses of expression restricted to S/G2/M, which are correlated between homologous promoters present in the same cell. The analysis of static, single-cell mRNA measurements at different points along the cell cycle corroborates these findings. Our results demonstrate that highly variable mRNA distributions in yeast are not solely the result of randomly switching between periods of active and inactive gene expression, but instead largely driven by differences in transcriptional activity between G1 and S/G2/M.},
author = {Zopf, Cristopher J. and Quinn, Katie and Zeidman, Joshua and Maheshri, Narendra},
doi = {10.1371/journal.pcbi.1003161},
file = {:Users/nils/Documents/Mendeley Desktop/journal.pcbi.1003161.PDF:PDF},
isbn = {1553-7358 (Electronic)$\backslash$n1553-734X (Linking)},
issn = {1553734X},
journal = {PLoS Computational Biology},
number = {7},
pages = {1--12},
pmid = {23935476},
title = {{Cell-Cycle Dependence of Transcription Dominates Noise in Gene Expression}},
volume = {9},
year = {2013}
}
@article{Iwamoto2016,
abstract = {Cellular heterogeneity, which plays an essential role in biological phenomena, such as drug resistance and migration, is considered to arise from intrinsic (i.e., reaction kinetics) and extrinsic (i.e., protein variability) noise in the cell. However, the mechanistic effects of these types of noise to determine the heterogeneity of signal responses have not been elucidated. Here, we report that the output of epidermal growth factor (EGF) signaling activity is modulated by cellular noise, particularly by extrinsic noise of particular signaling components in the pathway. We developed a mathematical model of the EGF signaling pathway incorporating regulation between extracellular signal-regulated kinase (ERK) and nuclear pore complex (NPC), which is necessary for switch-like activation of the nuclear ERK response. As the threshold of switch-like behavior is more sensitive to perturbations than the graded response, the effect of biological noise is potentially critical for cell fate decision. Our simulation analysis indicated that extrinsic noise, but not intrinsic noise, contributes to cell-to-cell heterogeneity of nuclear ERK. In addition, we accurately estimated variations in abundance of the signal proteins between individual cells by direct comparison of experimental data with simulation results using Apparent Measurement Error (AME). AME was constant regardless of whether the protein levels varied in a correlated manner, while covariation among proteins influenced cell-to-cell heterogeneity of nuclear ERK, suppressing the variation. Simulations using the estimated protein abundances showed that each protein species has different effects on cell-to-cell variation in the nuclear ERK response. In particular, variability of EGF receptor, Ras, Raf, and MEK strongly influenced cellular heterogeneity, while others did not. Overall, our results indicated that cellular heterogeneity in response to EGF is strongly driven by extrinsic noise, and that such heterogeneity results from variability of particular protein species that function as sensitive nodes, which may contribute to the pathogenesis of human diseases.},
author = {Iwamoto, Kazunari and Shindo, Yuki and Takahashi, Koichi},
doi = {10.1371/journal.pcbi.1005222},
file = {:Users/nils/Documents/Mendeley Desktop/Iwamoto, Shindo, Takahashi - 2016 - Modeling Cellular Noise Underlying Heterogeneous Cell Responses in the Epidermal Growth Factor Signa.pdf:pdf},
issn = {15537358},
journal = {PLoS Computational Biology},
number = {11},
pages = {1--18},
pmid = {27902699},
title = {{Modeling Cellular Noise Underlying Heterogeneous Cell Responses in the Epidermal Growth Factor Signaling Pathway}},
volume = {12},
year = {2016}
}
@article{Kiviet2014,
abstract = {Elucidating the role of molecular stochasticity in cellular growth is central to understanding phenotypic heterogeneity and the stability of cellular proliferation. The inherent stochasticity of metabolic reaction events should have negligible effect, because of averaging over the many reaction events contributing to growth. Indeed, metabolism and growth are often considered to be constant for fixed conditions. Stochastic fluctuations in the expression level of metabolic enzymes could produce variations in the reactions they catalyse. However, whether such molecular fluctuations can affect growth is unclear, given the various stabilizing regulatory mechanisms, the slow adjustment of key cellular components such as ribosomes, and the secretion and buffering of excess metabolites. Here we use time-lapse microscopy to measure fluctuations in the instantaneous growth rate of single cells of Escherichia coli, and quantify time-resolved cross-correlations with the expression of lac genes and enzymes in central metabolism. We show that expression fluctuations of catabolically active enzymes can propagate and cause growth fluctuations, with transmission depending on the limitation of the enzyme to growth. Conversely, growth fluctuations propagate back to perturb expression. Accordingly, enzymes were found to transmit noise to other unrelated genes via growth. Homeostasis is promoted by a noise-cancelling mechanism that exploits fluctuations in the dilution of proteins by cell-volume expansion. The results indicate that molecular noise is propagated not only by regulatory proteins but also by metabolic reactions. They also suggest that cellular metabolism is inherently stochastic, and a generic source of phenotypic heterogeneity.},
author = {Kiviet, Daniel J. and Nghe, Philippe and Walker, Noreen and Boulineau, Sarah and Sunderlikova, Vanda and Tans, Sander J.},
doi = {10.1038/nature13582},
file = {:Users/nils/Documents/Mendeley Desktop/Kiviet et al. - 2014 - Stochasticity of metabolism and growth at the single-cell level.pdf:pdf},
isbn = {1476-4687 (Electronic)$\backslash$r0028-0836 (Linking)},
issn = {0028-0836},
journal = {Nature},
number = {7522},
pages = {376--379},
pmid = {25186725},
publisher = {Nature Publishing Group},
title = {{Stochasticity of metabolism and growth at the single-cell level}},
url = {http://www.nature.com/doifinder/10.1038/nature13582},
volume = {514},
year = {2014}
}
@article{Faure2017,
abstract = {Isogenic cells in a common environment show substantial cell-to-cell variation in gene expression, often referred to as “expression noise.” Here, we use multiple single-cell RNA-sequencing datasets to identify features associated with high or low expression noise in mouse embryonic stem cells. These include the core promoter architecture of a gene, with CpG island promoters and a TATA box associated with low and high noise, respectively. High noise is also associated with “conflicting” chromatin states—the absence of transcription-associated histone modifications or the presence of repressive ones in active genes. Genes regulated by pluripotency factors through super-enhancers show high and correlated expression variability, consistent with fluctuations in the pluripotent state. Together, our results provide an integrated view of how core promoters, chromatin, regulation, and pluripotency fluctuations contribute to the variability of gene expression across individual stem cells. An integrative computational analysis identifies regulatory features that are statistically associated with high and low gene expression noise in mouse embryonic stem cells.},
author = {Faure, Andre J. and Schmiedel, J{\"{o}}rn M. and Lehner, Ben},
doi = {10.1016/j.cels.2017.10.003},
file = {:Users/nils/Documents/Mendeley Desktop/Faure, Schmiedel, Lehner - 2017 - Systematic Analysis of the Determinants of Gene Expression Noise in Embryonic Stem Cells.pdf:pdf},
issn = {24054720},
journal = {Cell Systems},
keywords = {chromatin,dosage compensation,embryonic stem cells,noise,promoter architectures,transcription},
number = {5},
pages = {471--484},
pmid = {29102610},
title = {{Systematic Analysis of the Determinants of Gene Expression Noise in Embryonic Stem Cells}},
volume = {5},
year = {2017}
}
@article{Morgan2018,
abstract = {Population phenotypic variation can arise from genetic differences between individuals, or from cellular heterogeneity in an isogenic group of cells or organisms. The emergence of gene expression differences between genetically identical cells is referred to as gene expression noise, the sources of which are not well understood. In this work, by studying gene expression noise between multiple cell lineages and mammalian species, we find consistent evidence of a role for CpG islands as sources of gene expression noise. Variation in noise among CpG island promoters can be partially attributed to differences in island size, in which short islands have noisier gene expression. Building on these findings, we investigate the potential for short CpG islands to act as fast response elements to environmental stimuli. Specifically, we find that these islands are enriched amongst primary response genes in SWI/SNF-independent stimuli, suggesting that expression noise is an indicator of promoter responsiveness. Thus, through the integration of single-cell RNA expression profiling, chromatin landscape and temporal gene expression dynamics, we have uncovered a role for short CpG island promoters as fast response elements.},
author = {Morgan, Michael D. and Marioni, John C.},
doi = {10.1186/s13059-018-1461-x},
file = {:Users/nils/Documents/Mendeley Desktop/Morgan, Marioni - 2018 - CpG island composition differences are a source of gene expression noise indicative of promoter responsiveness.pdf:pdf},
issn = {1474760X},
journal = {Genome Biology},
keywords = {Gene expression noise,Promoter response,Single cell},
number = {1},
pages = {1--13},
pmid = {29945659},
publisher = {Genome Biology},
title = {{CpG island composition differences are a source of gene expression noise indicative of promoter responsiveness}},
volume = {19},
year = {2018}
}
@article{Ecker2017a,
author = {Ecker, Simone and Pancaldi, Vera and Valencia, Alfonso and Beck, Stephan and Paul, Dirk S.},
doi = {10.1002/bies.201700148},
file = {:Users/nils/Documents/Mendeley Desktop/Ecker et al. - 2017 - Epigenetic and Transcriptional Variability Shape Phenotypic Plasticity.pdf:pdf},
issn = {02659247},
journal = {BioEssays},
pages = {1700148},
pmid = {29251357},
title = {{Epigenetic and Transcriptional Variability Shape Phenotypic Plasticity}},
url = {http://doi.wiley.com/10.1002/bies.201700148},
year = {2017}
}
@article{Vallejos2016,
author = {Vallejos, Catalina A and Richardson, Sylvia and Marioni, John C},
doi = {10.1101/035949},
file = {:Users/nils/Documents/Mendeley Desktop/art{\%}3A10.1186{\%}2Fs13059-016-0930-3.pdf:pdf},
journal = {Genome Biology},
keywords = {cellular heterogeneity,differential expression,single-cell rna-seq},
number = {70},
title = {{Beyond comparisons of means: understanding changes in gene expression at the single-cell level}},
url = {http://biorxiv.org/content/early/2016/01/05/035949.abstract},
volume = {17},
year = {2016}
}
@article{Eling2018,
author = {Eling, Nils and Richard, Arianne C. and Richardson, Sylvia and Marioni, John C. and Vallejos, Catalina A.},
doi = {10.1016/j.cels.2018.06.011},
file = {:Users/nils/Documents/Mendeley Desktop/PIIS2405471218302783 (1).pdf:pdf},
issn = {24054712},
journal = {Cell Systems},
number = {3},
pages = {1--11},
pmid = {30172840},
publisher = {Elsevier Inc.},
title = {{Correcting the Mean-Variance Dependency for Differential Variability Testing Using Single-Cell RNA Sequencing Data}},
url = {https://linkinghub.elsevier.com/retrieve/pii/S2405471218302783},
volume = {7},
year = {2018}
}
@article{Martinez-jimenez2017,
author = {Martinez-Jimenez, Celia P. and Eling, Nils and Chen, Hung-Chang and Vallejos, Catalina A and Kolodziejczyk, Aleksandra A and Connor, Frances and Stojic, Lovorka and Rayner, Timothy F and Stubbington, Michael J T and Teichmann, Sarah A and de la Roche, Maike and Marioni, John C and Odom, Duncan T},
doi = {10.1126/science.aah4115},
file = {:Users/nils/Documents/Mendeley Desktop/1433.full.pdf:pdf},
journal = {Science},
pages = {1433--1436},
title = {{Aging increases cell-to-cell transcriptional variability upon immune stimulation}},
volume = {1436},
year = {2017}
}
@incollection{Kim2019,
abstract = {Single-cell RNA-seq (scRNA-seq) provides a comprehensive measurement of stochasticity in transcription, but the limitations of the technology have prevented its application to dissect variability in RNA processing events such as splicing. In this chapter, we review the challenges in splicing isoform quantification in scRNA-seq data and discuss BRIE (Bayesian regression for isoform estimation), a recently proposed Bayesian hierarchical model which resolves these problems by learning an informative prior distribution from sequence features. We illustrate the usage of BRIE with a case study on 130 mouse cells during gastrulation.},
author = {Kim, Beomseok and Lee, Eunmin and Kim, Jong Kyoung},
booktitle = {Computational Methods for Single-Cell Data Analysis},
doi = {10.1007/978-1-4939-9057-3},
file = {:Users/nils/Documents/Mendeley Desktop/2019{\_}Book{\_}ComputationalMethodsForSingle-.pdf:pdf},
isbn = {978-1-4939-9056-6},
issn = {1940-6029},
keywords = {Alternative splicing,Bayesian model,Isoform quantification,Single-cell RNA-seq},
pages = {25--43},
pmid = {30758827},
title = {{Analysis of Technical and Biological Variabilityin Single-Cell RNA Sequencing}},
url = {http://www.ncbi.nlm.nih.gov/pubmed/30758827{\%}0Ahttp://link.springer.com/10.1007/978-1-4939-9057-3{\_}12{\%}0Ahttp://link.springer.com/10.1007/978-1-4939-9057-3},
volume = {1935},
year = {2019}
}
@article{Lun2016pooling,
abstract = {Normalization of single-cell RNA sequencing data is necessary to eliminate cell-specific biases prior to downstream analyses. However, this is not straightforward for noisy single-cell data where many counts are zero. We present a novel approach where expression values are summed across pools of cells, and the summed values are used for normalization. Pool-based size factors are then deconvolved to yield cell-based factors. Our deconvolution approach outperforms existing methods for accurate normalization of cell-specific biases in simulated data. Similar behavior is observed in real data, where deconvolution improves the relevance of results of downstream analyses.},
author = {Lun, Aaron T. L. and Bach, Karsten and Marioni, John C.},
doi = {10.1186/s13059-016-0947-7},
file = {:Users/nils/Documents/Mendeley Desktop/L. Lun, Bach, Marioni - 2016 - Pooling across cells to normalize single-cell RNA sequencing data with many zero counts.pdf:pdf},
issn = {1474-760X},
journal = {Genome Biology},
keywords = {differential expression,normalization,single-cell rna-seq},
number = {1},
pages = {75},
pmid = {27122128},
publisher = {Genome Biology},
title = {{Pooling across cells to normalize single-cell RNA sequencing data with many zero counts}},
url = {http://genomebiology.biomedcentral.com/articles/10.1186/s13059-016-0947-7},
volume = {17},
year = {2016}
}
@article{Kolodziejczyk2015cell,
author = {Kolodziejczyk, Aleksandra A. and Kim, Jong Kyoung and Tsang, Jason C.H. and Ilicic, Tomislav and Henriksson, Johan and Natarajan, Kedar N. and Tuck, Alex C. and Gao, Xuefei and B{\"{u}}hler, Marc and Liu, Pentao and Marioni, John C. and Teichmann, Sarah A.},
doi = {10.1016/j.stem.2015.09.011},
file = {:Users/nils/Documents/Mendeley Desktop/Kolodziejczyk et al. - 2015 - Single cell RNA-sequencing of pluripotent states unlocks modular transcriptional variation.pdf:pdf},
issn = {19345909},
journal = {Cell Stem Cell},
pages = {471--485},
title = {{Single cell RNA-sequencing of pluripotent states unlocks modular transcriptional variation}},
url = {http://linkinghub.elsevier.com/retrieve/pii/S193459091500418X},
volume = {17},
year = {2015}
}
@article{Young2010,
abstract = {We present GOseq, an application for performing Gene Ontology (GO) analysis on RNA-seq data. GO analysis is widely used to reduce complexity and highlight biological processes in genome-wide expression studies, but standard methods give biased results on RNA-seq data due to over-detection of differential expression for long and highly expressed transcripts. Application of GOseq to a prostate cancer data set shows that GOseq dramatically changes the results, highlighting categories more consistent with the known biology.},
author = {Young, Matthew D and Wakefield, Matthew J and Smyth, Gordon K and Oshlack, Alicia},
file = {:Users/nils/Documents/Mendeley Desktop/document (3).pdf:pdf},
journal = {Genome Biology},
title = {{Gene ontology analysis for RNA-seq: accounting for selection bias}},
url = {http://genomebiology.com/2010/11/2/R14},
volume = {11},
year = {2010}
}
@article{Durinck2005,
abstract = {biomaRt is a new Bioconductor package that integrates BioMart data resources with data analysis software in Bioconductor. It can annotate a wide range of gene or gene product identifiers (e.g. Entrez-Gene and Affymetrix probe identifiers) with information such as gene symbol, chromosomal coordinates, Gene Ontology and OMIM annotation. Furthermore biomaRt enables retrieval of genomic sequences and single nucleotide polymorphism information, which can be used in data analysis. Fast and up-to-date data retrieval is possible as the package executes direct SQL queries to the BioMart databases (e.g. Ensembl). The biomaRt package provides a tight integration of large, public or locally installed BioMart databases with data analysis in Bioconductor creating a powerful environment for biological data mining.},
author = {Durinck, Steffen and Moreau, Yves and Kasprzyk, Arek and Davis, Sean and {De Moor}, Bart and Brazma, Alvis and Huber, Wolfgang},
doi = {10.1093/bioinformatics/bti525},
file = {:Users/nils/Documents/Mendeley Desktop/bti525.pdf:pdf},
issn = {13674803},
journal = {Bioinformatics},
number = {16},
pages = {3439--3440},
title = {{BioMart and Bioconductor: A powerful link between biological databases and microarray data analysis}},
volume = {21},
year = {2005}
}
@article{Ilicic2016,
author = {Ilicic, Tomislav and Kim, Jong Kyoung and Kolodziejczyk, Aleksandra A and Bagger, Frederik Otzen and Mccarthy, Davis James and Marioni, John C and Teichmann, Sarah A},
doi = {10.1186/s13059-016-0888-1},
file = {:Users/nils/Documents/Mendeley Desktop/s13059-016-0888-1.pdf:pdf},
isbn = {1305901608881},
issn = {1474-760X},
journal = {Genome Biology},
number = {29},
pages = {1--15},
publisher = {Genome Biology},
title = {{Classification of low quality cells from single-cell RNA-seq data}},
url = {http://dx.doi.org/10.1186/s13059-016-0888-1},
volume = {17},
year = {2016}
}
@article{Vallejos2017,
abstract = {Single-cell transcriptomics is becoming an important component of the molecular biologist's toolkit. A critical step when analyzing data generated using this technology is normalization. However, normalization is typically performed using methods developed for bulk RNA sequencing or even microarray data, and the suitability of these methods for single-cell transcriptomics has not been assessed. We here discuss commonly used normalization approaches and illustrate how these can produce misleading results. Finally, we present alternative approaches and provide recommendations for single-cell RNA sequencing users.},
author = {Vallejos, Catalina A and Risso, Davide and Scialdone, Antonio and Dudoit, Sandrine and Marioni, John C},
doi = {10.1038/nmeth.4292},
file = {:Users/nils/Documents/Mendeley Desktop/vallejos2017.pdf:pdf},
issn = {1548-7091},
journal = {Nature Methods},
number = {6},
pages = {565--571},
pmid = {28504683},
publisher = {Nature Publishing Group},
title = {{Normalizing single-cell RNA sequencing data: challenges and opportunities}},
url = {http://www.nature.com/doifinder/10.1038/nmeth.4292},
volume = {14},
year = {2017}
}
@article{Shalek2014,
author = {Shalek, Alex K and Satija, Rahul and Shuga, Joe and Trombetta, John J and Gennert, Dave and Lu, Diana and Chen, Peilin and Gertner, Rona S and Gaublomme, Jellert T and Yosef, Nir and Schwartz, Schraga and Fowler, Brian and Weaver, Suzanne and Wang, Jing and Wang, Xiaohui and Ding, Ruihua and Raychowdhury, Raktima and Friedman, Nir and Hacohen, Nir and Park, Hongkun and May, Andrew P and Regev, Aviv},
doi = {10.1038/nature13437},
file = {:Users/nils/Documents/Mendeley Desktop/Shalek{\_}2014{\_}Nature13437.pdf:pdf},
issn = {0028-0836},
journal = {Nature},
number = {7505},
pages = {263--269},
publisher = {Nature Publishing Group},
title = {{Single-cell RNA-seq reveals dynamic paracrine control of cellular variation}},
url = {http://dx.doi.org/10.1038/nature13437},
volume = {510},
year = {2014}
}
@article{Antolovic2017,
author = {Antolovi{\'{c}}, Vlatka and Miermont, Agnes and Corrigan, Adam M. and Chubb, Jonathan R.},
doi = {10.1016/j.cub.2017.05.028},
file = {:Users/nils/Documents/Mendeley Desktop/PIIS096098221730564X.pdf:pdf},
issn = {09609822},
journal = {Current Biology},
pages = {1811--1817},
title = {{Generation of Single-Cell Transcript Variability by Repression}},
url = {http://linkinghub.elsevier.com/retrieve/pii/S096098221730564X},
volume = {27},
year = {2017}
}
@article{Borroto2000,
author = {Jose, Ester San and Borroto, Aldo and Niedergang, Florence and Alcover, Andres and Alarcon, Balbino},
file = {:Users/nils/Documents/Mendeley Desktop/1-s2.0-S1074761300801697-main.pdf:pdf},
journal = {Immunity},
pages = {161--170},
title = {{Triggering the TCR complexes causes the downregulation of nonengaged receptors by signal transduction-dependent mechanism}},
volume = {12},
year = {2000}
}
@article{Araki2017,
abstract = {Translation is a critical process in protein synthesis, but translational regulation in antigen-specific T cells in vivo has not been well defined. Here we have characterized the translatome of virus-specific CD8(+) effector T cells (Teff cells) during acute infection of mice with lymphocytic choriomeningitis virus (LCMV). Antigen-specific T cells exerted dynamic translational control of gene expression that correlated with cell proliferation and stimulation via the T cell antigen receptor (TCR). The translation of mRNAs that encode translation machinery, including ribosomal proteins, was upregulated during the T cell clonal-expansion phase, followed by inhibition of the translation of those transcripts when the CD8(+) Teff cells stopped dividing just before the contraction phase. That translational suppression was more pronounced in terminal effector cells than in memory precursor cells and was regulated by antigenic stimulation and signals from the kinase mTOR. Our studies show that translation of transcripts encoding ribosomal proteins is regulated during the differentiation of CD8(+) Teff cells and might have a role in fate 'decisions' involved in the formation of memory cells.},
author = {Araki, Koichi and Morita, Masahiro and Bederman, Annelise G. and Konieczny, Bogumila T. and Kissick, Haydn T. and Sonenberg, Nahum and Ahmed, Rafi},
doi = {10.1038/ni.3795},
file = {:Users/nils/Documents/Mendeley Desktop/araki2017.pdf:pdf},
issn = {15292916},
journal = {Nature Immunology},
number = {9},
pages = {1046--1057},
pmid = {28714979},
publisher = {Nature Publishing Group},
title = {{Translation is actively regulated during the differentiation of CD8 + effector T cells}},
url = {http://dx.doi.org/10.1038/ni.3795},
volume = {18},
year = {2017}
}
@article{Tan2017,
abstract = {Global transcriptomic and proteomic analyses of T cells have been rich sources of unbiased data for understanding T-cell activation. Lack of full concordance of these datasets has illustrated that important facets of T-cell activation are controlled at the level of translation. We undertook translatome analysis of CD8 T-cell activation, combining polysome profiling and microarray analysis. We revealed that altering T-cell receptor stimulation influenced recruitment of mRNAs to heavy polysomes and translation of subsets of genes. A major pathway that was compromised, when TCR signaling was suboptimal, was linked to ribosome biogenesis, a rate-limiting factor in both cell growth and proliferation. Defective TCR signaling affected transcription and processing of ribosomal RNA precursors, as well as the translation of specific ribosomal proteins and translation factors. Mechanistically, IL-2 production was compromised in weakly stimulated T cells, affecting the abundance of Myc protein, a known regulator of ribosome biogenesis. Consequently, weakly activated T cells showed impaired production of ribosomes and a failure to maintain proliferative capacity after stimulation. We demonstrate that primary T cells respond to various environmental cues by regulating ribosome biogenesis and mRNA translation at multiple levels to sustain proliferation and differentiation.},
author = {Tan, Thomas C. J. and Knight, John and Sbarrato, Thomas and Dudek, Kate and Willis, Anne E. and Zamoyska, Rose},
doi = {10.1073/pnas.1700939114},
file = {:Users/nils/Documents/Mendeley Desktop/Tan et al. - 2017 - Suboptimal T-cell receptor signaling compromises protein translation, ribosome biogenesis, and proliferation of mous.pdf:pdf},
isbn = {1700939114},
issn = {0027-8424},
journal = {Proceedings of the National Academy of Sciences},
number = {30},
pages = {E6117--E6126},
pmid = {28696283},
title = {{Suboptimal T-cell receptor signaling compromises protein translation, ribosome biogenesis, and proliferation of mouse CD8 T cells}},
url = {http://www.pnas.org/lookup/doi/10.1073/pnas.1700939114},
volume = {114},
year = {2017}
}
@article{Hagai2018,
author = {Hagai, Tzachi and Chen, Xi and Miragaia, Ricardo J and Rostom, Raghd and Gomes, Tom{\'{a}}s and Kunowska, Natalia and Henriksson, Johan and Park, Jong-eun and Proserpio, Valentina and Donati, Giacomo and Bossini-castillo, Lara and Braga, Felipe A Vieira and Naamati, Guy and Fletcher, James and Stephenson, Emily and Vegh, Peter and Trynka, Gosia and Kondova, Ivanela and Dennis, Mike and Haniffa, Muzlifah and Nourmohammad, Armita and L{\"{a}}ssig, Michael and Teichmann, Sarah A.},
doi = {10.1038/s41586-018-0657-2},
file = {:Users/nils/Documents/Mendeley Desktop/hagai2018.pdf:pdf},
journal = {Nature},
pages = {197--202},
title = {{Gene expression variability across cells and species shapes innate immunity}},
volume = {563},
year = {2018}
}
@article{Klein2015,
author = {Klein, Allon M. and Mazutis, Linas and Akartuna, Ilke and Tallapragada, Naren and Veres, Adrian and Li, Victor and Peshkin, Leonid and Weitz, David A. and Kirschner, Marc W.},
doi = {10.1016/j.cell.2015.04.044},
file = {:Users/nils/Documents/Mendeley Desktop/Klein et al. - 2015 - Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells.pdf:pdf},
issn = {00928674},
journal = {Cell},
number = {5},
pages = {1187--1201},
publisher = {Elsevier Inc.},
title = {{Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells}},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0092867415005000},
volume = {161},
year = {2015}
}
@article{Kernfeld2018,
abstract = {Thymus development is critical to the adaptive immune system, yet a comprehensive transcriptional framework capturing thymus organogenesis at single-cell resolution is still needed. We applied single-cell RNA sequencing (RNA-seq) to capture 8 days of thymus development, perturbations of T cell receptor rearrangement, and in vitro organ cultures, producing profiles of 24,279 cells. We resolved transcriptional heterogeneity of developing lymphocytes, and genetic perturbation confirmed T cell identity of conventional and non-conventional lymphocytes. We characterized maturation dynamics of thymic epithelial cells in vivo, classified cell maturation state in a thymic organ culture, and revealed the intrinsic capacity of thymic epithelium to preserve transcriptional regularity despite exposure to exogenous retinoic acid. Finally, by integrating the cell atlas with human genome-wide association study (GWAS) data and autoimmune-disease-related genes, we implicated embryonic thymus-resident cells as possible participants in autoimmune disease etiologies. This resource provides a single-cell transcriptional framework for biological discovery and molecular analysis of thymus organogenesis. Studies of thymus development typically lack single-cell resolution, use indirect readouts, or target narrow cell subsets. Using single-cell RNA sequencing during thymus organogenesis, Kernfeld and Genga et al. reveal cellular heterogeneity, interrogate developmental dynamics by direct observation, and pinpoint cell-specific expression patterns in stromal and blood populations.},
author = {Kernfeld, Eric M. and Genga, Ryan M.J. and Neherin, Kashfia and Magaletta, Margaret E. and Xu, Ping and Maehr, Ren{\'{e}}},
doi = {10.1016/j.immuni.2018.04.015},
file = {:Users/nils/Documents/Mendeley Desktop/[email protected]:pdf},
isbn = {1074-7613},
issn = {10974180},
journal = {Immunity},
keywords = {Drop-seq,cell atlas,development,lymphocytes,lymphoid organ,single-cell RNA-seq,thymic epithelium,thymus,thymus organogenesis,transcriptomics},
pages = {1--13},
pmid = {29884461},
title = {{A Single-Cell Transcriptomic Atlas of Thymus Organogenesis Resolves Cell Types and Developmental Maturation}},
volume = {48},
year = {2018}
}
@book{Carroll1998,
address = {Chichester, UK},
author = {Carroll, Raymond J.},
booktitle = {Encyclopedia of Biostatistics},
doi = {10.1002/9781118445112.stat05178},
file = {:Users/nils/Documents/Mendeley Desktop/Carroll, Carroll, J. - 2014 - Measurement Error in Epidemiologic Studies.pdf:pdf},
keywords = {SIMEX (simulation extrapolation),attenuation,errors‐in‐variables,instrumental variables,latent variables,measurement error,misclassification,regression calibration},
month = {dec},
publisher = {John Wiley {\&} Sons, Ltd},
title = {{Measurement Error in Epidemiologic Studies}},
year = {1998}
}
@article{Macosko2015,
author = {Macosko, Evan Z. and Basu, Anindita and Satija, Rahul and Nemesh, James and Shekhar, Karthik and Goldman, Melissa and Tirosh, Itay and Bialas, Allison R. and Kamitaki, Nolan and Martersteck, Emily M. and Trombetta, John J. and Weitz, David A. and Sanes, Joshua R. and Shalek, Alex K. and Regev, Aviv and McCarroll, Steven A.},
doi = {10.1016/j.cell.2015.05.002},
file = {:Users/nils/Documents/Mendeley Desktop/Macosko et al. - 2015 - Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets.pdf:pdf},
issn = {00928674},
journal = {Cell},
number = {5},
pages = {1202--1214},
publisher = {Elsevier},
title = {{Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets}},
url = {http://linkinghub.elsevier.com/retrieve/pii/S0092867415005498},
volume = {161},
year = {2015}
}
@article{Saelens2019,
title = {A Comparison of Single-Cell Trajectory Inference Methods},
author = {Saelens, Wouter and Cannoodt, Robrecht and Todorov, Helena and Saeys, Yvan},
year = {2019},
month = may,
volume = {37},
pages = {547--554},
issn = {1087-0156, 1546-1696},
doi = {10.1038/s41587-019-0071-9},
file = {/home/alan/Zotero/storage/YZ3DGBAE/Saelens et al. - 2019 - A comparison of single-cell trajectory inference m.pdf},
journal = {Nature Biotechnology},
language = {en},
number = {5}
}
@article{Smith1993,
title = {Bayesian {{Computation Via}} the {{Gibbs Sampler}} and {{Related Markov Chain Monte Carlo Methods}}},
author = {Smith, A. F. M. and Roberts, G. O.},
year = {1993},
month = sep,
volume = {55},
pages = {3--23},
issn = {00359246},
doi = {10.1111/j.2517-6161.1993.tb01466.x},
abstract = {The use of the Gibbs sampler for Bayesian computation is reviewed and illustrated in the context of some canonical examples. Other Markov chain Monte Carlo simulation methods are also briefly described, and comments are made on the advantages of sample-based approaches for Bayesian inference summaries.},
file = {/home/alan/Zotero/storage/W9RP9PJS/Smith and Roberts - 1993 - Bayesian Computation Via the Gibbs Sampler and Rel.pdf},
journal = {Journal of the Royal Statistical Society: Series B (Methodological)},
language = {en},
number = {17}
}
@article{Cowles1996,
title = {Markov {{Chain Monte Carlo Convergence Diagnostics}}: {{A Comparative Review}}},
shorttitle = {Markov {{Chain Monte Carlo Convergence Diagnostics}}},
author = {Cowles, Mary Kathryn and Carlin, Bradley P.},
year = {1996},
month = jun,
volume = {91},
pages = {883--904},
issn = {0162-1459, 1537-274X},
doi = {10.1080/01621459.1996.10476956},
file = {/home/alan/Zotero/storage/HI2LZNDX/Cowles and Carlin - 1996 - Markov Chain Monte Carlo Convergence Diagnostics .pdf},
journal = {Journal of the American Statistical Association},
language = {en},
number = {434}
}
@article{Eling2019,
title = {Challenges in Measuring and Understanding Biological Noise},
author = {Eling, Nils and Morgan, Michael D. and Marioni, John C.},
year = {2019},
month = sep,
volume = {20},
pages = {536--548},
issn = {1471-0056, 1471-0064},
doi = {10.1038/s41576-019-0130-6},
abstract = {Biochemical reactions are intrinsically stochastic, leading to variation in the production of mRNAs and proteins within cells. In the scientific literature, this source of variation is typically referred to as `noise'. The observed variability in molecular phenotypes arises from a combination of processes that amplify and attenuate noise. Our ability to quantify cell-t o-cell variability in numerous biological contexts has been revolutionized by recent advances in single-c ell technology, from imaging approaches through to `omics' strategies. However, defining, accurately measuring and disentangling the stochastic and deterministic components of cell-to-cell variability is challenging. In this Review, we discuss the sources, impact and function of molecular phenotypic variability and highlight future directions to understand its role.},
file = {/home/alan/Zotero/storage/X26SMRRG/Eling et al. - 2019 - Challenges in measuring and understanding biologic.pdf},
journal = {Nature Reviews Genetics},
language = {en},
number = {9}
}
@article{Townes2019,
title = {Feature Selection and Dimension Reduction for Single-Cell {{RNA}}-{{Seq}} Based on a Multinomial Model},
author = {Townes, F. William and Hicks, Stephanie C. and Aryee, Martin J. and Irizarry, Rafael A.},
year = {2019},
month = dec,
volume = {20},
pages = {295},
issn = {1474-760X},
doi = {10.1186/s13059-019-1861-6},
abstract = {Single-cell RNA-Seq (scRNA-Seq) profiles gene expression of individual cells. Recent scRNA-Seq datasets have incorporated unique molecular identifiers (UMIs). Using negative controls, we show UMI counts follow multinomial sampling with no zero inflation. Current normalization procedures such as log of counts per million and feature selection by highly variable genes produce false variability in dimension reduction. We propose simple multinomial methods, including generalized principal component analysis (GLM-PCA) for non-normal distributions, and feature selection using deviance. These methods outperform the current practice in a downstream clustering assessment using ground truth datasets.},
file = {/home/alan/Zotero/storage/DVCYIZEA/Townes et al. - 2019 - Feature selection and dimension reduction for sing.pdf},
journal = {Genome Biology},
language = {en},
number = {1}
}
@article{Lopez2018,
title = {Deep Generative Modeling for Single-Cell Transcriptomics},
author = {Lopez, Romain and Regier, Jeffrey and Cole, Michael B. and Jordan, Michael I. and Yosef, Nir},
year = {2018},
month = dec,
volume = {15},
pages = {1053--1058},
issn = {1548-7091, 1548-7105},
doi = {10.1038/s41592-018-0229-2},
file = {/home/alan/Zotero/storage/ZGXHGR5W/Lopez et al. - 2018 - Deep generative modeling for single-cell transcrip.pdf},
journal = {Nature Methods},
language = {en},
number = {12}
}
@techreport{Amezquita2019,
title = {Orchestrating {{Single}}-{{Cell Analysis}} with {{Bioconductor}}},
author = {Amezquita, Robert A. and Carey, Vince J. and Carpp, Lindsay N. and Geistlinger, Ludwig and Lun, Aaron T. L. and Marini, Federico and {Rue-Albrecht}, Kevin and Risso, Davide and Soneson, Charlotte and Waldron, Levi and Pag{\`e}s, Herv{\'e} and Smith, Mike and Huber, Wolfgang and Morgan, Martin and Gottardo, Raphael and Hicks, Stephanie C.},
year = {2019},
month = mar,
institution = {{Genomics}},