-
Notifications
You must be signed in to change notification settings - Fork 0
/
Copy patharticles.bib
3373 lines (3167 loc) · 345 KB
/
articles.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
@article{aagaardPlacentaHarborsUnique2014,
title = {The Placenta Harbors a Unique Microbiome},
author = {Aagaard, Kjersti and Ma, Jun and Antony, Kathleen M. and Ganu, Radhika and Petrosino, Joseph and Versalovic, James},
year = {2014},
month = may,
volume = {6},
pages = {237ra65},
issn = {1946-6242},
doi = {10.1126/scitranslmed.3008599},
abstract = {Humans and their microbiomes have coevolved as a physiologic community composed of distinct body site niches with metabolic and antigenic diversity. The placental microbiome has not been robustly interrogated, despite recent demonstrations of intracellular bacteria with diverse metabolic and immune regulatory functions. A population-based cohort of placental specimens collected under sterile conditions from 320 subjects with extensive clinical data was established for comparative 16S ribosomal DNA-based and whole-genome shotgun (WGS) metagenomic studies. Identified taxa and their gene carriage patterns were compared to other human body site niches, including the oral, skin, airway (nasal), vaginal, and gut microbiomes from nonpregnant controls. We characterized a unique placental microbiome niche, composed of nonpathogenic commensal microbiota from the Firmicutes, Tenericutes, Proteobacteria, Bacteroidetes, and Fusobacteria phyla. In aggregate, the placental microbiome profiles were most akin (Bray-Curtis dissimilarity {$<$}0.3) to the human oral microbiome. 16S-based operational taxonomic unit analyses revealed associations of the placental microbiome with a remote history of antenatal infection (permutational multivariate analysis of variance, P = 0.006), such as urinary tract infection in the first trimester, as well as with preterm birth {$<$}37 weeks (P = 0.001).},
journal = {Science Translational Medicine},
keywords = {菌群发育,里程碑},
language = {eng},
number = {237},
pmcid = {PMC4929217},
pmid = {24848255}
}
@article{adamGrowingTreeArchaea2017,
title = {The Growing Tree of {{Archaea}}: New Perspectives on Their Diversity, Evolution and Ecology},
shorttitle = {The Growing Tree of {{Archaea}}},
author = {Adam, Panagiotis S. and Borrel, Guillaume and {Brochier-Armanet}, C{\'e}line and Gribaldo, Simonetta},
year = {2017},
month = nov,
volume = {11},
pages = {2407--2425},
issn = {1751-7370},
doi = {10.1038/ismej.2017.122},
abstract = {The Archaea occupy a key position in the Tree of Life, and are a major fraction of microbial diversity. Abundant in soils, ocean sediments and the water column, they have crucial roles in processes mediating global carbon and nutrient fluxes. Moreover, they represent an important component of the human microbiome, where their role in health and disease is still unclear. The development of culture-independent sequencing techniques has provided unprecedented access to genomic data from a large number of so far inaccessible archaeal lineages. This is revolutionizing our view of the diversity and metabolic potential of the Archaea in a wide variety of environments, an important step toward understanding their ecological role. The archaeal tree is being rapidly filled up with new branches constituting phyla, classes and orders, generating novel challenges for high-rank systematics, and providing key information for dissecting the origin of this domain, the evolutionary trajectories that have shaped its current diversity, and its relationships with Bacteria and Eukarya. The present picture is that of a huge diversity of the Archaea, which we are only starting to explore.},
journal = {The ISME journal},
keywords = {古菌,里程碑},
language = {eng},
number = {11},
pmcid = {PMC5649171},
pmid = {28777382}
}
@article{ahnHumanGutMicrobiome2013,
title = {Human {{Gut Microbiome}} and {{Risk}} for {{Colorectal Cancer}}},
author = {Ahn, Jiyoung and Sinha, Rashmi and Pei, Zhiheng and Dominianni, Christine and Wu, Jing and Shi, Jianxin and Goedert, James J. and Hayes, Richard B. and Yang, Liying},
year = {2013},
month = dec,
volume = {105},
pages = {1907--1911},
publisher = {{Oxford Academic}},
issn = {0027-8874},
doi = {10.1093/jnci/djt300},
abstract = {Abstract. We tested the hypothesis that an altered community of gut microbes is associated with risk of colorectal cancer (CRC) in a study of 47 CRC case subj},
journal = {JNCI: Journal of the National Cancer Institute},
language = {en},
number = {24}
}
@article{arpaiaMetabolitesProducedCommensal2013,
title = {Metabolites Produced by Commensal Bacteria Promote Peripheral Regulatory {{T}}-Cell Generation},
author = {Arpaia, Nicholas and Campbell, Clarissa and Fan, Xiying and Dikiy, Stanislav and {van der Veeken}, Joris and {deRoos}, Paul and Liu, Hui and Cross, Justin R. and Pfeffer, Klaus and Coffer, Paul J. and Rudensky, Alexander Y.},
year = {2013},
month = dec,
volume = {504},
pages = {451--455},
publisher = {{Nature Publishing Group}},
issn = {1476-4687},
doi = {10.1038/nature12726},
abstract = {In mice, provision of butyrate\textemdash a short-chain fatty acid produced by commensal microorganisms during starch fermentation\textemdash facilitates extrathymic generation and differentiation of Foxp3+ regulatory T cells, demonstrating that metabolic by-products are sensed by cells of the immune system and affect the balance between pro- and anti-inflammatory cells.},
copyright = {2013 Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
journal = {Nature},
language = {en},
number = {7480}
}
@article{arumugamEnterotypesHumanGut2011,
title = {Enterotypes of the Human Gut Microbiome},
author = {Arumugam, Manimozhiyan and Raes, Jeroen and Pelletier, Eric and Le Paslier, Denis and Yamada, Takuji and Mende, Daniel R. and Fernandes, Gabriel R. and Tap, Julien and Bruls, Thomas and Batto, Jean-Michel and Bertalan, Marcelo and Borruel, Natalia and Casellas, Francesc and Fernandez, Leyden and Gautier, Laurent and Hansen, Torben and Hattori, Masahira and Hayashi, Tetsuya and Kleerebezem, Michiel and Kurokawa, Ken and Leclerc, Marion and Levenez, Florence and Manichanh, Chaysavanh and Nielsen, H. Bj{\o}rn and Nielsen, Trine and Pons, Nicolas and Poulain, Julie and Qin, Junjie and {Sicheritz-Ponten}, Thomas and Tims, Sebastian and Torrents, David and Ugarte, Edgardo and Zoetendal, Erwin G. and Wang, Jun and Guarner, Francisco and Pedersen, Oluf and {de Vos}, Willem M. and Brunak, S{\o}ren and Dor{\'e}, Joel and Weissenbach, Jean and Ehrlich, S. Dusko and Bork, Peer},
year = {2011},
month = may,
volume = {473},
pages = {174--180},
publisher = {{Nature Publishing Group}},
issn = {1476-4687},
doi = {10.1038/nature09944},
abstract = {The human gut microbiota consists of a huge number of species and varies greatly between individuals. A comparative metagenomic analysis of the human gut microbiomes of 39 individuals from 6 countries shows that despite this diversity, the microbiota composition can be classified into at least 3 distinct groups, or enterotypes. The enterotypes contain functional markers that correlate with individual features such as age and body mass index, a feature that may be of use in the diagnosis of numerous human disorders such as colorectal cancer and diabetes.},
copyright = {2011 Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
journal = {Nature},
keywords = {菌群稳定性,里程碑},
language = {en},
number = {7346}
}
@article{ashManipulatingMicrobiota2016,
title = {Manipulating the {{Microbiota}}},
author = {Ash, C. and Mueller, K.},
year = {2016},
month = apr,
volume = {352},
pages = {530--531},
issn = {0036-8075, 1095-9203},
doi = {10.1126/science.352.6285.530},
journal = {Science},
language = {en},
number = {6285}
}
@article{atarashiInductionColonicRegulatory2011,
title = {Induction of {{Colonic Regulatory T Cells}} by {{Indigenous Clostridium Species}}},
author = {Atarashi, Koji and Tanoue, Takeshi and Shima, Tatsuichiro and Imaoka, Akemi and Kuwahara, Tomomi and Momose, Yoshika and Cheng, Genhong and Yamasaki, Sho and Saito, Takashi and Ohba, Yusuke and Taniguchi, Tadatsugu and Takeda, Kiyoshi and Hori, Shohei and Ivanov, Ivaylo I. and Umesaki, Yoshinori and Itoh, Kikuji and Honda, Kenya},
year = {2011},
month = jan,
volume = {331},
pages = {337--341},
publisher = {{American Association for the Advancement of Science}},
issn = {0036-8075, 1095-9203},
doi = {10.1126/science.1198469},
abstract = {CD4+ T regulatory cells (Tregs), which express the Foxp3 transcription factor, play a critical role in the maintenance of immune homeostasis. Here, we show that in mice, Tregs were most abundant in the colonic mucosa. The spore-forming component of indigenous intestinal microbiota, particularly clusters IV and XIVa of the genus Clostridium, promoted Treg cell accumulation. Colonization of mice by a defined mix of Clostridium strains provided an environment rich in transforming growth factor\textendash{$\beta$} and affected Foxp3+ Treg number and function in the colon. Oral inoculation of Clostridium during the early life of conventionally reared mice resulted in resistance to colitis and systemic immunoglobulin E responses in adult mice, suggesting a new therapeutic approach to autoimmunity and allergy. Bacteria of the genus Clostridium promote the induction of suppressor T cells in the colons of mice. Bacteria of the genus Clostridium promote the induction of suppressor T cells in the colons of mice.},
chapter = {Report},
copyright = {Copyright \textcopyright{} 2011, American Association for the Advancement of Science},
journal = {Science},
language = {en},
number = {6015},
pmid = {21205640}
}
@article{backhedDynamicsStabilizationHuman2015,
title = {Dynamics and {{Stabilization}} of the {{Human Gut Microbiome}} during the {{First Year}} of {{Life}}},
author = {B{\"a}ckhed, Fredrik and Roswall, Josefine and Peng, Yangqing and Feng, Qiang and Jia, Huijue and {Kovatcheva-Datchary}, Petia and Li, Yin and Xia, Yan and Xie, Hailiang and Zhong, Huanzi and Khan, Muhammad Tanweer and Zhang, Jianfeng and Li, Junhua and Xiao, Liang and {Al-Aama}, Jumana and Zhang, Dongya and Lee, Ying Shiuan and Kotowska, Dorota and Colding, Camilla and Tremaroli, Valentina and Yin, Ye and Bergman, Stefan and Xu, Xun and Madsen, Lise and Kristiansen, Karsten and Dahlgren, Jovanna and Wang, Jun},
year = {2015},
month = may,
volume = {17},
pages = {690--703},
issn = {19313128},
doi = {10.1016/j.chom.2015.04.004},
journal = {Cell Host \& Microbe},
language = {en},
number = {5}
}
@article{backhedGutMicrobiotaEnvironmental2004,
title = {The Gut Microbiota as an Environmental Factor That Regulates Fat Storage},
author = {B{\"a}ckhed, Fredrik and Ding, Hao and Wang, Ting and Hooper, Lora V. and Koh, Gou Young and Nagy, Andras and Semenkovich, Clay F. and Gordon, Jeffrey I.},
year = {2004},
month = nov,
volume = {101},
pages = {15718--15723},
publisher = {{National Academy of Sciences}},
issn = {0027-8424, 1091-6490},
doi = {10.1073/pnas.0407076101},
abstract = {New therapeutic targets for noncognitive reductions in energy intake, absorption, or storage are crucial given the worldwide epidemic of obesity. The gut microbial community (microbiota) is essential for processing dietary polysaccharides. We found that conventionalization of adult germ-free (GF) C57BL/6 mice with a normal microbiota harvested from the distal intestine (cecum) of conventionally raised animals produces a 60\% increase in body fat content and insulin resistance within 14 days despite reduced food intake. Studies of GF and conventionalized mice revealed that the microbiota promotes absorption of monosaccharides from the gut lumen, with resulting induction of de novo hepatic lipogenesis. Fasting-induced adipocyte factor (Fiaf), a member of the angiopoietin-like family of proteins, is selectively suppressed in the intestinal epithelium of normal mice by conventionalization. Analysis of GF and conventionalized, normal and Fiaf knockout mice established that Fiaf is a circulating lipoprotein lipase inhibitor and that its suppression is essential for the microbiota-induced deposition of triglycerides in adipocytes. Studies of Rag1-/- animals indicate that these host responses do not require mature lymphocytes. Our findings suggest that the gut microbiota is an important environmental factor that affects energy harvest from the diet and energy storage in the host.},
chapter = {Biological Sciences},
copyright = {Copyright \textcopyright{} 2004, The National Academy of Sciences. Freely available online through the PNAS open access option.},
journal = {Proceedings of the National Academy of Sciences},
language = {en},
number = {44},
pmid = {15505215}
}
@article{backhedMechanismsUnderlyingResistance2007,
title = {Mechanisms Underlying the Resistance to Diet-Induced Obesity in Germ-Free Mice},
author = {B{\"a}ckhed, Fredrik and Manchester, Jill K. and Semenkovich, Clay F. and Gordon, Jeffrey I.},
year = {2007},
month = jan,
volume = {104},
pages = {979--984},
issn = {0027-8424},
doi = {10.1073/pnas.0605374104},
abstract = {The trillions of microbes that colonize our adult intestines function collectively as a metabolic organ that communicates with, and complements, our own human metabolic apparatus. Given the worldwide epidemic in obesity, there is interest in how interactions between human and microbial metabolomes may affect our energy balance. Here we report that, in contrast to mice with a gut microbiota, germ-free (GF) animals are protected against the obesity that develops after consuming a Western-style, high-fat, sugar-rich diet. Their persistently lean phenotype is associated with increased skeletal muscle and liver levels of phosphorylated AMP-activated protein kinase (AMPK) and its downstream targets involved in fatty acid oxidation (acetylCoA carboxylase; carnitine-palmitoyltransferase). Moreover, GF knockout mice lacking fasting-induced adipose factor (Fiaf), a circulating lipoprotein lipase inhibitor whose expression is normally selectively suppressed in the gut epithelium by the microbiota, are not protected from diet-induced obesity. Although GF Fiaf-/- animals exhibit similar levels of phosphorylated AMPK as their wild-type littermates in liver and gastrocnemius muscle, they have reduced expression of genes encoding the peroxisomal proliferator-activated receptor coactivator (Pgc-1alpha) and enzymes involved in fatty acid oxidation. Thus, GF animals are protected from diet-induced obesity by two complementary but independent mechanisms that result in increased fatty acid metabolism: (i) elevated levels of Fiaf, which induces Pgc-1alpha; and (ii) increased AMPK activity. Together, these findings support the notion that the gut microbiota can influence both sides of the energy balance equation, and underscore the importance of considering our metabolome in a supraorganismal context.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
language = {eng},
number = {3},
pmcid = {PMC1764762},
pmid = {17210919}
}
@article{bashirTolllikeReceptorSignaling2004,
title = {Toll-like Receptor 4 Signaling by Intestinal Microbes Influences Susceptibility to Food Allergy},
author = {Bashir, Mohamed Elfatih H. and Louie, Steve and Shi, Hai Ning and {Nagler-Anderson}, Cathryn},
year = {2004},
month = jun,
volume = {172},
pages = {6978--6987},
issn = {0022-1767},
doi = {10.4049/jimmunol.172.11.6978},
abstract = {The mechanisms by which signaling by the innate immune system controls susceptibility to allergy are poorly understood. In this report, we show that intragastric administration of a food allergen with a mucosal adjuvant induces allergen-specific IgE, elevated plasma histamine levels, and anaphylactic symptoms in three different strains of mice lacking a functional receptor for bacterial LPS (Toll-like receptor 4 (TLR4)), but not in MHC-matched or congenic controls. Susceptibility to allergy correlates with a Th2-biased cytokine response in both the mucosal (mesenteric lymph node and Peyer's patch) and systemic (spleen) tissues of TLR4-mutant or -deficient mice. TLR4-mutant mice are not inherently impaired in their ability to regulate Th1 cytokine production because they respond to stimulation via TLR9. Coadministration of CpG oligodeoxynucleotides during sensitization of TLR4-mutant mice with allergen plus CT abrogates anaphylactic symptoms and Ag-specific IgE, and results in a Th1-polarized cytokine response. When the composition of the bacterial flora is reduced and altered by antibiotic administration (beginning at 2 wk of age), TLR4 wild-type mice become as susceptible to the induction of allergy as their TLR4-mutant counterparts. Both allergen-specific IgE and Th2 cytokine responses are reduced in antibiotic-treated mice in which the flora has been allowed to repopulate. Taken together, our results suggest that TLR4-dependent signals provided by the intestinal commensal flora inhibit the development of allergic responses to food Ags.},
journal = {Journal of Immunology (Baltimore, Md.: 1950)},
keywords = {粘膜免疫,里程碑},
language = {eng},
number = {11},
pmid = {15153518}
}
@article{bikMolecularAnalysisBacterial2006,
title = {Molecular Analysis of the Bacterial Microbiota in the Human Stomach},
author = {Bik, Elisabeth M. and Eckburg, Paul B. and Gill, Steven R. and Nelson, Karen E. and Purdom, Elizabeth A. and Francois, Fritz and {Perez-Perez}, Guillermo and Blaser, Martin J. and Relman, David A.},
year = {2006},
month = jan,
volume = {103},
pages = {732--737},
issn = {0027-8424},
doi = {10.1073/pnas.0506655103},
abstract = {The microbiota of the human stomach and the influence of Helicobacter pylori colonization on its composition remain largely unknown. We characterized bacterial diversity within the human gastric mucosa by using a small subunit 16S rDNA clone library approach and analyzed 1,833 sequences generated by broad-range bacterial PCR from 23 gastric endoscopic biopsy samples. A diverse community of 128 phylotypes was identified, featuring diversity at this site greater than previously described. The majority of sequences were assigned to the Proteobacteria, Firmicutes, Actinobacteria, Bacteroidetes, and Fusobacteria phyla. Ten percent of the phylotypes were previously uncharacterized, including a Deinococcus-related organism, relatives of which have been found in extreme environments but not reported before in humans. The gastric clone libraries from 19 subjects contained H. pylori rDNA; however, only 12 of these subjects tested positive for H. pylori by conventional laboratory methods. Statistical analysis revealed a large degree of intersubject variability of the gastric ecosystem. The presence of H. pylori did not affect the composition of the gastric community. This gastric bacterial rDNA data set was significantly different from sequence collections of the human mouth and esophagus described in other studies, indicating that the human stomach may be home to a distinct microbial eco-system. The gastric microbiota may play important, as-yet-undiscovered roles in human health and disease.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
keywords = {技术进步,里程碑},
language = {eng},
number = {3},
pmcid = {PMC1334644},
pmid = {16407106}
}
@article{blaserAntibioticUseIts2016,
title = {Antibiotic Use and Its Consequences for the Normal Microbiome},
author = {Blaser, M. J.},
year = {2016},
month = apr,
volume = {352},
pages = {544--545},
issn = {0036-8075, 1095-9203},
doi = {10.1126/science.aad9358},
journal = {Science},
language = {en},
number = {6285}
}
@article{bokulichAntibioticsBirthMode2016,
title = {Antibiotics, Birth Mode, and Diet Shape Microbiome Maturation during Early Life},
author = {Bokulich, Nicholas A. and Chung, Jennifer and Battaglia, Thomas and Henderson, Nora and Jay, Melanie and Li, Huilin and D Lieber, Arnon and Wu, Fen and {Perez-Perez}, Guillermo I. and Chen, Yu and Schweizer, William and Zheng, Xuhui and Contreras, Monica and {Dominguez-Bello}, Maria Gloria and Blaser, Martin J.},
year = {2016},
month = jun,
volume = {8},
pages = {343ra82},
issn = {1946-6242},
doi = {10.1126/scitranslmed.aad7121},
abstract = {Early childhood is a critical stage for the foundation and development of both the microbiome and host. Early-life antibiotic exposures, cesarean section, and formula feeding could disrupt microbiome establishment and adversely affect health later in life. We profiled microbial development during the first 2 years of life in a cohort of 43 U.S. infants and identified multiple disturbances associated with antibiotic exposures, cesarean section, and formula feeding. These exposures contributed to altered establishment of maternal bacteria, delayed microbiome development, and altered {$\alpha$}-diversity. These findings illustrate the complexity of early-life microbiome development and its sensitivity to perturbation.},
journal = {Science Translational Medicine},
keywords = {菌群发育,里程碑},
language = {eng},
number = {343},
pmcid = {PMC5308924},
pmid = {27306664}
}
@article{bondarRegulationMucosalImmunity2019,
title = {Regulation of Mucosal Immunity by the Microbiota},
author = {Bondar, Tanya},
year = {2019},
month = jun,
publisher = {{Nature Publishing Group}},
doi = {10.1038/d42859-019-00014-2},
abstract = {Discover the world's best science and medicine | Nature.com},
copyright = {2019 Nature},
journal = {Nature Research},
keywords = {粘膜免疫,里程碑},
language = {en}
}
@article{borrelGenomicsMetagenomicsTrimethylamineutilizing2017,
title = {Genomics and Metagenomics of Trimethylamine-Utilizing {{Archaea}} in the Human Gut Microbiome},
author = {Borrel, Guillaume and McCann, Angela and Deane, Jennifer and Neto, Marta C. and Lynch, Denise B. and Brug{\`e}re, Jean-Fran{\c c}ois and O'Toole, Paul W.},
year = {2017},
month = sep,
volume = {11},
pages = {2059--2074},
issn = {1751-7370},
doi = {10.1038/ismej.2017.72},
abstract = {The biological significance of Archaea in the human gut microbiota is largely unclear. We recently reported genomic and biochemical analyses of the Methanomassiliicoccales, a novel order of methanogenic Archaea dwelling in soil and the animal digestive tract. We now show that these Methanomassiliicoccales are present in published microbiome data sets from eight countries. They are represented by five Operational Taxonomic Units present in at least four cohorts and phylogenetically distributed into two clades. Genes for utilizing trimethylamine (TMA), a bacterial precursor to an atherosclerogenic human metabolite, were present in four of the six novel Methanomassiliicoccales genomes assembled from ELDERMET metagenomes. In addition to increased microbiota TMA production capacity in long-term residential care subjects, abundance of TMA-utilizing Methanomassiliicoccales correlated positively with bacterial gene count for TMA production and negatively with fecal TMA concentrations. The two large Methanomassiliicoccales clades have opposite correlations with host health status in the ELDERMET cohort and putative distinct genomic signatures for gut adaptation.},
journal = {The ISME journal},
keywords = {古菌,里程碑},
language = {eng},
number = {9},
pmcid = {PMC5563959},
pmid = {28585938}
}
@article{brunelloGutMicrobiotaTransfer2019,
title = {Gut Microbiota Transfer Experiments in Germ-Free Animals},
author = {Brunello, Lucia},
year = {2019},
month = jun,
publisher = {{Nature Publishing Group}},
doi = {10.1038/d42859-019-00009-z},
abstract = {Discover the world's best science and medicine | Nature.com},
copyright = {2019 Nature},
journal = {Nature Research},
keywords = {无菌动物,里程碑},
language = {en}
}
@article{caniChangesGutMicrobiota2008,
title = {Changes in {{Gut Microbiota Control Metabolic Endotoxemia}}-{{Induced Inflammation}} in {{High}}-{{Fat Diet}}\textendash{{Induced Obesity}} and {{Diabetes}} in {{Mice}}},
author = {Cani, Patrice D. and Bibiloni, Rodrigo and Knauf, Claude and Waget, Aur{\'e}lie and Neyrinck, Audrey M. and Delzenne, Nathalie M. and Burcelin, R{\'e}my},
year = {2008},
month = jun,
volume = {57},
pages = {1470--1481},
publisher = {{American Diabetes Association}},
issn = {0012-1797, 1939-327X},
doi = {10.2337/db07-1403},
abstract = {OBJECTIVE\textemdash Diabetes and obesity are characterized by a low-grade inflammation whose molecular origin is unknown. We previously determined, first, that metabolic endotoxemia controls the inflammatory tone, body weight gain, and diabetes, and second, that high-fat feeding modulates gut microbiota and the plasma concentration of lipopolysaccharide (LPS), i.e., metabolic endotoxemia. Therefore, it remained to demonstrate whether changes in gut microbiota control the occurrence of metabolic diseases. RESEARCH DESIGN AND METHODS\textemdash We changed gut microbiota by means of antibiotic treatment to demonstrate, first, that changes in gut microbiota could be responsible for the control of metabolic endotoxemia, the low-grade inflammation, obesity, and type 2 diabetes and, second, to provide some mechanisms responsible for such effect. RESULTS\textemdash We found that changes of gut microbiota induced by an antibiotic treatment reduced metabolic endotoxemia and the cecal content of LPS in both high-fat\textendash fed and ob/ob mice. This effect was correlated with reduced glucose intolerance, body weight gain, fat mass development, lower inflammation, oxidative stress, and macrophage infiltration marker mRNA expression in visceral adipose tissue. Importantly, high-fat feeding strongly increased intestinal permeability and reduced the expression of genes coding for proteins of the tight junctions. Furthermore, the absence of CD14 in ob/ob CD14-/- mutant mice mimicked the metabolic and inflammatory effects of antibiotics. CONCLUSIONS\textemdash This new finding demonstrates that changes in gut microbiota controls metabolic endotoxemia, inflammation, and associated disorders by a mechanism that could increase intestinal permeability. It would thus be useful to develop strategies for changing gut microbiota to control, intestinal permeability, metabolic endotoxemia, and associated disorders.},
chapter = {Metabolism},
copyright = {DIABETES},
journal = {Diabetes},
language = {en},
number = {6},
pmid = {18305141}
}
@article{caniChangesGutMicrobiota2009,
title = {Changes in Gut Microbiota Control Inflammation in Obese Mice through a Mechanism Involving {{GLP}}-2-Driven Improvement of Gut Permeability},
author = {Cani, Patrice D. and Possemiers, S. and de Wiele, T. Van and Guiot, Y. and Everard, A. and Rottier, O. and Geurts, L. and Naslain, D. and Neyrinck, A. and Lambert, D. M. and Muccioli, G. G. and Delzenne, N. M.},
year = {2009},
month = aug,
volume = {58},
pages = {1091--1103},
publisher = {{BMJ Publishing Group}},
issn = {0017-5749, 1468-3288},
doi = {10.1136/gut.2008.165886},
abstract = {Background and aims: Obese and diabetic mice display enhanced intestinal permeability and metabolic endotoxaemia that participate in the occurrence of metabolic disorders. Our recent data support the idea that a selective increase of Bifidobacterium spp. reduces the impact of high-fat diet-induced metabolic endotoxaemia and inflammatory disorders. Here, we hypothesised that prebiotic modulation of gut microbiota lowers intestinal permeability, by a mechanism involving glucagon-like peptide-2 (GLP-2) thereby improving inflammation and metabolic disorders during obesity and diabetes. Methods: Study 1: ob/ob mice (Ob-CT) were treated with either prebiotic (Ob-Pre) or non-prebiotic carbohydrates as control (Ob-Cell). Study 2: Ob-CT and Ob-Pre mice were treated with GLP-2 antagonist or saline. Study 3: Ob-CT mice were treated with a GLP-2 agonist or saline. We assessed changes in the gut microbiota, intestinal permeability, gut peptides, intestinal epithelial tight-junction proteins ZO-1 and occludin (qPCR and immunohistochemistry), hepatic and systemic inflammation. Results: Prebiotic-treated mice exhibited a lower plasma lipopolysaccharide (LPS) and cytokines, and a decreased hepatic expression of inflammatory and oxidative stress markers. This decreased inflammatory tone was associated with a lower intestinal permeability and improved tight-junction integrity compared to controls. Prebiotic increased the endogenous intestinotrophic proglucagon-derived peptide (GLP-2) production whereas the GLP-2 antagonist abolished most of the prebiotic effects. Finally, pharmacological GLP-2 treatment decreased gut permeability, systemic and hepatic inflammatory phenotype associated with obesity to a similar extent as that observed following prebiotic-induced changes in gut microbiota. Conclusion: We found that a selective gut microbiota change controls and increases endogenous GLP-2 production, and consequently improves gut barrier functions by a GLP-2-dependent mechanism, contributing to the improvement of gut barrier functions during obesity and diabetes.},
chapter = {Inflammatory bowel disease},
copyright = {\textcopyright{} Cani et al 2009. This is an open-access article distributed under the terms of the Creative Commons Attribution Non-commercial License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.},
journal = {Gut},
language = {en},
number = {8},
pmid = {19240062}
}
@article{caniMetabolicEndotoxemiaInitiates2007,
title = {Metabolic {{Endotoxemia Initiates Obesity}} and {{Insulin Resistance}}},
author = {Cani, Patrice D. and Amar, Jacques and Iglesias, Miguel Angel and Poggi, Marjorie and Knauf, Claude and Bastelica, Delphine and Neyrinck, Audrey M. and Fava, Francesca and Tuohy, Kieran M. and Chabo, Chantal and Waget, Aur{\'e}lie and Delm{\'e}e, Evelyne and Cousin, B{\'e}atrice and Sulpice, Thierry and Chamontin, Bernard and Ferri{\`e}res, Jean and Tanti, Jean-Fran{\c c}ois and Gibson, Glenn R. and Casteilla, Louis and Delzenne, Nathalie M. and Alessi, Marie Christine and Burcelin, R{\'e}my},
year = {2007},
month = jul,
volume = {56},
pages = {1761--1772},
publisher = {{American Diabetes Association}},
issn = {0012-1797, 1939-327X},
doi = {10.2337/db06-1491},
abstract = {Diabetes and obesity are two metabolic diseases characterized by insulin resistance and a low-grade inflammation. Seeking an inflammatory factor causative of the onset of insulin resistance, obesity, and diabetes, we have identified bacterial lipopolysaccharide (LPS) as a triggering factor. We found that normal endotoxemia increased or decreased during the fed or fasted state, respectively, on a nutritional basis and that a 4-week high-fat diet chronically increased plasma LPS concentration two to three times, a threshold that we have defined as metabolic endotoxemia. Importantly, a high-fat diet increased the proportion of an LPS-containing microbiota in the gut. When metabolic endotoxemia was induced for 4 weeks in mice through continuous subcutaneous infusion of LPS, fasted glycemia and insulinemia and whole-body, liver, and adipose tissue weight gain were increased to a similar extent as in high-fat\textendash fed mice. In addition, adipose tissue F4/80-positive cells and markers of inflammation, and liver triglyceride content, were increased. Furthermore, liver, but not whole-body, insulin resistance was detected in LPS-infused mice. CD14 mutant mice resisted most of the LPS and high-fat diet\textendash induced features of metabolic diseases. This new finding demonstrates that metabolic endotoxemia dysregulates the inflammatory tone and triggers body weight gain and diabetes. We conclude that the LPS/CD14 system sets the tone of insulin sensitivity and the onset of diabetes and obesity. Lowering plasma LPS concentration could be a potent strategy for the control of metabolic diseases.},
chapter = {Obesity Studies},
copyright = {DIABETES},
journal = {Diabetes},
language = {en},
number = {7},
pmid = {17456850}
}
@article{caniSelectiveIncreasesBifidobacteria2007,
title = {Selective Increases of Bifidobacteria in Gut Microflora Improve High-Fat-Diet-Induced Diabetes in Mice through a Mechanism Associated with Endotoxaemia},
author = {Cani, Patrice D. and Neyrinck, A. M. and Fava, F. and Knauf, C. and Burcelin, R. G. and Tuohy, K. M. and Gibson, G. R. and Delzenne, N. M.},
year = {2007},
month = nov,
volume = {50},
pages = {2374--2383},
issn = {1432-0428},
doi = {10.1007/s00125-007-0791-0},
abstract = {Recent evidence suggests that a particular gut microbial community may favour occurrence of the metabolic diseases. Recently, we reported that high-fat (HF) feeding was associated with higher endotoxaemia and lower Bifidobacterium species (spp.) caecal content in mice. We therefore tested whether restoration of the quantity of caecal Bifidobacterium spp. could modulate metabolic endotoxaemia, the inflammatory tone and the development of diabetes.},
journal = {Diabetologia},
language = {en},
number = {11}
}
@article{caporasoMovingPicturesHuman2011,
title = {Moving Pictures of the Human Microbiome},
author = {Caporaso, J. Gregory and Lauber, Christian L. and Costello, Elizabeth K. and {Berg-Lyons}, Donna and Gonzalez, Antonio and Stombaugh, Jesse and Knights, Dan and Gajer, Pawel and Ravel, Jacques and Fierer, Noah and Gordon, Jeffrey I. and Knight, Rob},
year = {2011},
volume = {12},
pages = {R50},
issn = {1474-760X},
doi = {10.1186/gb-2011-12-5-r50},
abstract = {BACKGROUND: Understanding the normal temporal variation in the human microbiome is critical to developing treatments for putative microbiome-related afflictions such as obesity, Crohn's disease, inflammatory bowel disease and malnutrition. Sequencing and computational technologies, however, have been a limiting factor in performing dense time series analysis of the human microbiome. Here, we present the largest human microbiota time series analysis to date, covering two individuals at four body sites over 396 timepoints. RESULTS: We find that despite stable differences between body sites and individuals, there is pronounced variability in an individual's microbiota across months, weeks and even days. Additionally, only a small fraction of the total taxa found within a single body site appear to be present across all time points, suggesting that no core temporal microbiome exists at high abundance (although some microbes may be present but drop below the detection threshold). Many more taxa appear to be persistent but non-permanent community members. CONCLUSIONS: DNA sequencing and computational advances described here provide the ability to go beyond infrequent snapshots of our human-associated microbial ecology to high-resolution assessments of temporal variations over protracted periods, within and between body habitats and individuals. This capacity will allow us to define normal variation and pathologic states, and assess responses to therapeutic interventions.},
journal = {Genome Biology},
keywords = {菌群稳定性,里程碑},
language = {eng},
number = {5},
pmcid = {PMC3271711},
pmid = {21624126}
}
@article{cashSymbioticBacteriaDirect2006,
title = {Symbiotic Bacteria Direct Expression of an Intestinal Bactericidal Lectin},
author = {Cash, Heather L. and Whitham, Cecilia V. and Behrendt, Cassie L. and Hooper, Lora V.},
year = {2006},
month = aug,
volume = {313},
pages = {1126--1130},
issn = {1095-9203},
doi = {10.1126/science.1127119},
abstract = {The mammalian intestine harbors complex societies of beneficial bacteria that are maintained in the lumen with minimal penetration of mucosal surfaces. Microbial colonization of germ-free mice triggers epithelial expression of RegIIIgamma, a secreted C-type lectin. RegIIIgamma binds intestinal bacteria but lacks the complement recruitment domains present in other microbe-binding mammalian C-type lectins. We show that RegIIIgamma and its human counterpart, HIP/PAP, are directly antimicrobial proteins that bind their bacterial targets via interactions with peptidoglycan carbohydrate. We propose that these proteins represent an evolutionarily primitive form of lectin-mediated innate immunity, and that they reveal intestinal strategies for maintaining symbiotic host-microbial relationships.},
journal = {Science (New York, N.Y.)},
keywords = {粘膜免疫,里程碑},
language = {eng},
number = {5790},
pmcid = {PMC2716667},
pmid = {16931762}
}
@article{castellarinFusobacteriumNucleatumInfection2012,
title = {Fusobacterium Nucleatum Infection Is Prevalent in Human Colorectal Carcinoma},
author = {Castellarin, Mauro and Warren, Ren{\'e} L. and Freeman, J. Douglas and Dreolini, Lisa and Krzywinski, Martin and Strauss, Jaclyn and Barnes, Rebecca and Watson, Peter and {Allen-Vercoe}, Emma and Moore, Richard A. and Holt, Robert A.},
year = {2012},
month = jan,
volume = {22},
pages = {299--306},
publisher = {{Cold Spring Harbor Lab}},
issn = {1088-9051, 1549-5469},
doi = {10.1101/gr.126516.111},
abstract = {An estimated 15\% or more of the cancer burden worldwide is attributable to known infectious agents. We screened colorectal carcinoma and matched normal tissue specimens using RNA-seq followed by host sequence subtraction and found marked over-representation of Fusobacterium nucleatum sequences in tumors relative to control specimens. F. nucleatum is an invasive anaerobe that has been linked previously to periodontitis and appendicitis, but not to cancer. Fusobacteria are rare constituents of the fecal microbiota, but have been cultured previously from biopsies of inflamed gut mucosa. We obtained a Fusobacterium isolate from a frozen tumor specimen; this showed highest sequence similarity to a known gut mucosa isolate and was confirmed to be invasive. We verified overabundance of Fusobacterium sequences in tumor versus matched normal control tissue by quantitative PCR analysis from a total of 99 subjects (p = 2.5 \texttimes{} 10-6), and we observed a positive association with lymph node metastasis.},
journal = {Genome Research},
language = {en},
number = {2},
pmid = {22009989}
}
@misc{ChangDaoYuJun,
title = {肠{$\cdot$}道 | 于君:5400字详解大肠癌和菌群关系的5大问题!},
abstract = {肠道专业人士的招牌演讲,截止2019年8月,已有53位讲者在7期录制中奉献55个精彩演讲。},
howpublished = {https://www.mr-gut.cn/talks/s/ff4c145befbd4001b1ea59e7c75a367a}
}
@article{chenCiteSpaceIIDetecting2006,
title = {{{CiteSpace II}}: {{Detecting}} and Visualizing Emerging Trends and Transient Patterns in Scientific Literature},
shorttitle = {{{CiteSpace II}}},
author = {Chen, Chaomei},
year = {2006},
month = feb,
volume = {57},
pages = {359--377},
issn = {15322882, 15322890},
doi = {10.1002/asi.20317},
journal = {Journal of the American Society for Information Science and Technology},
language = {en},
number = {3}
}
@article{chuMaturationInfantMicrobiome2017,
title = {Maturation of the Infant Microbiome Community Structure and Function across Multiple Body Sites and in Relation to Mode of Delivery},
author = {Chu, Derrick M. and Ma, Jun and Prince, Amanda L. and Antony, Kathleen M. and Seferovic, Maxim D. and Aagaard, Kjersti M.},
year = {2017},
month = mar,
volume = {23},
pages = {314--326},
issn = {1546-170X},
doi = {10.1038/nm.4272},
abstract = {Human microbial communities are characterized by their taxonomic, metagenomic and metabolic diversity, which varies by distinct body sites and influences human physiology. However, when and how microbial communities within each body niche acquire unique taxonomical and functional signatures in early life remains underexplored. We thus sought to determine the taxonomic composition and potential metabolic function of the neonatal and early infant microbiota across multiple body sites and assess the effect of the mode of delivery and its potential confounders or modifiers. A cohort of pregnant women in their early third trimester (n = 81) were prospectively enrolled for longitudinal sampling through 6 weeks after delivery, and a second matched cross-sectional cohort (n = 81) was additionally recruited for sampling once at the time of delivery. Samples across multiple body sites, including stool, oral gingiva, nares, skin and vagina were collected for each maternal-infant dyad. Whole-genome shotgun sequencing and sequencing analysis of the gene encoding the 16S rRNA were performed to interrogate the composition and function of the neonatal and maternal microbiota. We found that the neonatal microbiota and its associated functional pathways were relatively homogeneous across all body sites at delivery, with the notable exception of the neonatal meconium. However, by 6 weeks after delivery, the infant microbiota structure and function had substantially expanded and diversified, with the body site serving as the primary determinant of the composition of the bacterial community and its functional capacity. Although minor variations in the neonatal (immediately at birth) microbiota community structure were associated with the cesarean mode of delivery in some body sites (oral gingiva, nares and skin; R2 = 0.038), this was not true for neonatal stool (meconium; Mann-Whitney P {$>$} 0.05), and there was no observable difference in community function regardless of delivery mode. For infants at 6 weeks of age, the microbiota structure and function had expanded and diversified with demonstrable body site specificity (P {$<$} 0.001, R2 = 0.189) but without discernable differences in community structure or function between infants delivered vaginally or by cesarean surgery (P = 0.057, R2 = 0.007). We conclude that within the first 6 weeks of life, the infant microbiota undergoes substantial reorganization, which is primarily driven by body site and not by mode of delivery.},
journal = {Nature Medicine},
keywords = {菌群发育,里程碑},
language = {eng},
number = {3},
pmcid = {PMC5345907},
pmid = {28112736}
}
@article{claessonGutMicrobiotaComposition2012,
title = {Gut Microbiota Composition Correlates with Diet and Health in the Elderly},
author = {Claesson, Marcus J. and Jeffery, Ian B. and Conde, Susana and Power, Susan E. and O'Connor, Eibhl{\'i}s M. and Cusack, Siobh{\'a}n and Harris, Hugh M. B. and Coakley, Mairead and Lakshminarayanan, Bhuvaneswari and O'Sullivan, Orla and Fitzgerald, Gerald F. and Deane, Jennifer and O'Connor, Michael and Harnedy, Norma and O'Connor, Kieran and O'Mahony, Denis and {van Sinderen}, Douwe and Wallace, Martina and Brennan, Lorraine and Stanton, Catherine and Marchesi, Julian R. and Fitzgerald, Anthony P. and Shanahan, Fergus and Hill, Colin and Ross, R. Paul and O'Toole, Paul W.},
year = {2012},
month = aug,
volume = {488},
pages = {178--184},
publisher = {{Nature Publishing Group}},
issn = {1476-4687},
doi = {10.1038/nature11319},
abstract = {Alterations in intestinal microbiota composition are associated with several chronic conditions, including obesity and inflammatory diseases. The microbiota of older people displays greater inter-individual variation than that of younger adults. Here we show that the faecal microbiota composition from 178 elderly subjects formed groups, correlating with residence location in the community, day-hospital, rehabilitation or in long-term residential care. However, clustering of subjects by diet separated them by the same residence location and microbiota groupings. The separation of microbiota composition significantly correlated with measures of frailty, co-morbidity, nutritional status, markers of inflammation and with metabolites in faecal water. The individual microbiota of people in long-stay care was significantly less diverse than that of community dwellers. Loss of community-associated microbiota correlated with increased frailty. Collectively, the data support a relationship between diet, microbiota and health status, and indicate a role for diet-driven microbiota alterations in varying rates of health decline upon ageing.},
copyright = {2012 Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
journal = {Nature},
language = {en},
number = {7410}
}
@article{clarkCulturingAnaerobes2019,
title = {Culturing Anaerobes},
author = {Clark, Hannah},
year = {2019},
month = jun,
doi = {10.1038/d42859-019-00007-1},
abstract = {Discover the world's best science and medicine | Nature.com},
copyright = {2019 Nature},
journal = {Nature Research},
language = {en}
}
@article{claytonPharmacometabonomicIdentificationSignificant2009,
title = {Pharmacometabonomic Identification of a Significant Host-Microbiome Metabolic Interaction Affecting Human Drug Metabolism},
author = {Clayton, T. Andrew and Baker, David and Lindon, John C. and Everett, Jeremy R. and Nicholson, Jeremy K.},
year = {2009},
month = aug,
volume = {106},
pages = {14728--14733},
issn = {1091-6490},
doi = {10.1073/pnas.0904489106},
abstract = {We provide a demonstration in humans of the principle of pharmacometabonomics by showing a clear connection between an individual's metabolic phenotype, in the form of a predose urinary metabolite profile, and the metabolic fate of a standard dose of the widely used analgesic acetaminophen. Predose and postdose urinary metabolite profiles were determined by (1)H NMR spectroscopy. The predose spectra were statistically analyzed in relation to drug metabolite excretion to detect predose biomarkers of drug fate and a human-gut microbiome cometabolite predictor was identified. Thus, we found that individuals having high predose urinary levels of p-cresol sulfate had low postdose urinary ratios of acetaminophen sulfate to acetaminophen glucuronide. We conclude that, in individuals with high bacterially mediated p-cresol generation, competitive O-sulfonation of p-cresol reduces the effective systemic capacity to sulfonate acetaminophen. Given that acetaminophen is such a widely used and seemingly well-understood drug, this finding provides a clear demonstration of the immense potential and power of the pharmacometabonomic approach. However, we expect many other sulfonation reactions to be similarly affected by competition with p-cresol and our finding also has important implications for certain diseases as well as for the variable responses induced by many different drugs and xenobiotics. We propose that assessing the effects of microbiome activity should be an integral part of pharmaceutical development and of personalized health care. Furthermore, we envisage that gut bacterial populations might be deliberately manipulated to improve drug efficacy and to reduce adverse drug reactions.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
keywords = {Acetaminophen,Administration; Oral,Adolescent,Adult,Analgesics; Non-Narcotic,Bacteria,Cresols,Gastrointestinal Tract,Host-Pathogen Interactions,Humans,Magnetic Resonance Spectroscopy,Male,Middle Aged,Sulfates,Sulfuric Acid Esters,Young Adult,药物代谢,里程碑},
language = {eng},
number = {34},
pmcid = {PMC2731842},
pmid = {19667173}
}
@article{costelloBacterialCommunityVariation2009,
title = {Bacterial Community Variation in Human Body Habitats across Space and Time},
author = {Costello, Elizabeth K. and Lauber, Christian L. and Hamady, Micah and Fierer, Noah and Gordon, Jeffrey I. and Knight, Rob},
year = {2009},
month = dec,
volume = {326},
pages = {1694--1697},
issn = {1095-9203},
doi = {10.1126/science.1177486},
abstract = {Elucidating the biogeography of bacterial communities on the human body is critical for establishing healthy baselines from which to detect differences associated with diseases. To obtain an integrated view of the spatial and temporal distribution of the human microbiota, we surveyed bacteria from up to 27 sites in seven to nine healthy adults on four occasions. We found that community composition was determined primarily by body habitat. Within habitats, interpersonal variability was high, whereas individuals exhibited minimal temporal variability. Several skin locations harbored more diverse communities than the gut and mouth, and skin locations differed in their community assembly patterns. These results indicate that our microbiota, although personalized, varies systematically across body habitats and time; such trends may ultimately reveal how microbiome changes cause or prevent disease.},
journal = {Science (New York, N.Y.)},
keywords = {菌群稳定性,里程碑},
language = {eng},
number = {5960},
pmcid = {PMC3602444},
pmid = {19892944}
}
@article{davidDietRapidlyReproducibly2014,
title = {Diet Rapidly and Reproducibly Alters the Human Gut Microbiome},
author = {David, Lawrence A. and Maurice, Corinne F. and Carmody, Rachel N. and Gootenberg, David B. and Button, Julie E. and Wolfe, Benjamin E. and Ling, Alisha V. and Devlin, A. Sloan and Varma, Yug and Fischbach, Michael A. and Biddinger, Sudha B. and Dutton, Rachel J. and Turnbaugh, Peter J.},
year = {2014},
month = jan,
volume = {505},
pages = {559--563},
issn = {0028-0836},
doi = {10.1038/nature12820},
abstract = {Long-term dietary intake influences the structure and activity of the trillions of microorganisms residing in the human gut, but it remains unclear how rapidly and reproducibly the human gut microbiome responds to short-term macronutrient change. Here we show that the short-term consumption of diets composed entirely of animal or plant products alters microbial community structure and overwhelms inter-individual differences in microbial gene expression. The animal-based diet increased the abundance of bile-tolerant microorganisms (Alistipes, Bilophila and Bacteroides) and decreased the levels of Firmicutes that metabolize dietary plant polysaccharides (Roseburia, Eubacterium rectale and Ruminococcus bromii). Microbial activity mirrored differences between herbivorous and carnivorous mammals, reflecting trade-offs between carbohydrate and protein fermentation. Foodborne microbes from both diets transiently colonized the gut, including bacteria, fungi and even viruses. Finally, increases in the abundance and activity of Bilophila wadsworthia on the animal-based diet support a link between dietary fat, bile acids and the outgrowth of microorganisms capable of triggering inflammatory bowel disease. In concert, these results demonstrate that the gut microbiome can rapidly respond to altered diet, potentially facilitating the diversity of human dietary lifestyles.},
copyright = {\textcopyright{} 2013 Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
file = {E\:\\zotero.storage.2016\\Nature\\2014\\DavidL et al_Nature_2014.pdf},
journal = {Nature},
language = {en},
number = {7484}
}
@article{desaiDietaryFiberDeprivedGut2016,
title = {A {{Dietary Fiber}}-{{Deprived Gut Microbiota Degrades}} the {{Colonic Mucus Barrier}} and {{Enhances Pathogen Susceptibility}}},
author = {Desai, Mahesh S. and Seekatz, Anna M. and Koropatkin, Nicole M. and Kamada, Nobuhiko and Hickey, Christina A. and Wolter, Mathis and Pudlo, Nicholas A. and Kitamoto, Sho and Terrapon, Nicolas and Muller, Arnaud and Young, Vincent B. and Henrissat, Bernard and Wilmes, Paul and Stappenbeck, Thaddeus S. and N{\'u}{\~n}ez, Gabriel and Martens, Eric C.},
year = {2016},
month = nov,
volume = {167},
pages = {1339-1353.e21},
issn = {1097-4172},
doi = {10.1016/j.cell.2016.10.043},
abstract = {Despite the accepted health benefits of consuming dietary fiber, little is known about the mechanisms by which fiber deprivation impacts the gut microbiota and alters disease risk. Using a gnotobiotic mouse model, in which animals were colonized with a synthetic human gut microbiota composed of fully sequenced commensal bacteria, we elucidated the functional interactions between dietary fiber, the gut microbiota, and the colonic mucus barrier, which serves as a primary defense against enteric pathogens. We show that during chronic or intermittent dietary fiber deficiency, the gut microbiota resorts to host-secreted mucus glycoproteins as a nutrient source, leading to erosion of the colonic mucus barrier. Dietary fiber deprivation, together with a fiber-deprived, mucus-eroding microbiota, promotes greater epithelial access and lethal colitis by the mucosal pathogen, Citrobacter rodentium. Our work reveals intricate pathways linking diet, the gut microbiome, and intestinal barrier dysfunction, which could be exploited to improve health using dietary therapeutics.},
journal = {Cell},
keywords = {喂养菌群,里程碑},
language = {eng},
number = {5},
pmcid = {PMC5131798},
pmid = {27863247}
}
@article{digiulioTemporalSpatialVariation2015,
title = {Temporal and Spatial Variation of the Human Microbiota during Pregnancy},
author = {DiGiulio, Daniel B. and Callahan, Benjamin J. and McMurdie, Paul J. and Costello, Elizabeth K. and Lyell, Deirdre J. and Robaczewska, Anna and Sun, Christine L. and Goltsman, Daniela S. A. and Wong, Ronald J. and Shaw, Gary and Stevenson, David K. and Holmes, Susan P. and Relman, David A.},
year = {2015},
month = sep,
volume = {112},
pages = {11060--11065},
issn = {1091-6490},
doi = {10.1073/pnas.1502875112},
abstract = {Despite the critical role of the human microbiota in health, our understanding of microbiota compositional dynamics during and after pregnancy is incomplete. We conducted a case-control study of 49 pregnant women, 15 of whom delivered preterm. From 40 of these women, we analyzed bacterial taxonomic composition of 3,767 specimens collected prospectively and weekly during gestation and monthly after delivery from the vagina, distal gut, saliva, and tooth/gum. Linear mixed-effects modeling, medoid-based clustering, and Markov chain modeling were used to analyze community temporal trends, community structure, and vaginal community state transitions. Microbiota community taxonomic composition and diversity remained remarkably stable at all four body sites during pregnancy (P {$>$} 0.05 for trends over time). Prevalence of a Lactobacillus-poor vaginal community state type (CST 4) was inversely correlated with gestational age at delivery (P = 0.0039). Risk for preterm birth was more pronounced for subjects with CST 4 accompanied by elevated Gardnerella or Ureaplasma abundances. This finding was validated with a set of 246 vaginal specimens from nine women (four of whom delivered preterm). Most women experienced a postdelivery disturbance in the vaginal community characterized by a decrease in Lactobacillus species and an increase in diverse anaerobes such as Peptoniphilus, Prevotella, and Anaerococcus species. This disturbance was unrelated to gestational age at delivery and persisted for up to 1 y. These findings have important implications for predicting premature labor, a major global health problem, and for understanding the potential impact of a persistent, altered postpartum microbiota on maternal health, including outcomes of pregnancies following short interpregnancy intervals.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
keywords = {菌群稳定性,里程碑},
language = {eng},
number = {35},
pmcid = {PMC4568272},
pmid = {26283357}
}
@article{dioneQuasiuniversalMediumBreak2016,
title = {A Quasi-Universal Medium to Break the Aerobic/Anaerobic Bacterial Culture Dichotomy in Clinical Microbiology},
author = {Dione, N. and Khelaifia, S. and La Scola, B. and Lagier, J. C. and Raoult, D.},
year = {2016},
month = jan,
volume = {22},
pages = {53--58},
issn = {1469-0691},
doi = {10.1016/j.cmi.2015.10.032},
abstract = {In the mid-19th century, the dichotomy between aerobic and anaerobic bacteria was introduced. Nevertheless, the aerobic growth of strictly anaerobic bacterial species such as Ruminococcus gnavus and Fusobacterium necrophorum, in a culture medium containing antioxidants, was recently demonstrated. We tested aerobically the culture of 623 bacterial strains from 276 bacterial species including 82 strictly anaerobic, 154 facultative anaerobic, 31 aerobic and nine microaerophilic bacterial species as well as ten fungi. The basic culture medium was based on Schaedler agar supplemented with 1 g/L ascorbic acid and 0.1 g/L glutathione (R-medium). We successively optimized this media, adding 0.4 g/L uric acid, using separate autoclaving of the component, or adding haemin 0.1 g/L or {$\alpha$}-ketoglutarate 2 g/L. In the basic medium, 237 bacterial species and ten fungal species grew but with no growth of 36 bacterial species, including 22 strict anaerobes. Adding uric acid allowed the growth of 14 further species including eight strict anaerobes, while separate autoclaving allowed the growth of all tested bacterial strains. To extend its potential use for fastidious bacteria, we added haemin for Haemophilus influenzae, Haemophilus parainfluenzae and Eikenella corrodens and {$\alpha$}-ketoglutarate for Legionella pneumophila. This medium allowed the growth of all tested strains with the exception of Mycobacterium tuberculosis and Mycobacterium bovis. Testing primoculture and more fastidious species will constitute the main work to be done, but R-medium coupled with a rapid identification method (matrix-assisted laser desorption/ionization time-of-flight mass spectrometry) will facilitate the anaerobic culture in clinical microbiology laboratories.},
journal = {Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases},
keywords = {Anaerobes,antioxidant,ascorbic acid,Bacteria; Aerobic,Bacteria; Anaerobic,Bacterial Infections,Bacteriological Techniques,Culture Media,glutathione,Humans,uric acid,培养组学,里程碑},
language = {eng},
number = {1},
pmid = {26577141}
}
@article{dominguez-belloDeliveryModeShapes2010,
title = {Delivery Mode Shapes the Acquisition and Structure of the Initial Microbiota across Multiple Body Habitats in Newborns},
author = {{Dominguez-Bello}, Maria G. and Costello, Elizabeth K. and Contreras, Monica and Magris, Magda and Hidalgo, Glida and Fierer, Noah and Knight, Rob},
year = {2010},
month = jun,
volume = {107},
pages = {11971--11975},
issn = {1091-6490},
doi = {10.1073/pnas.1002601107},
abstract = {Upon delivery, the neonate is exposed for the first time to a wide array of microbes from a variety of sources, including maternal bacteria. Although prior studies have suggested that delivery mode shapes the microbiota's establishment and, subsequently, its role in child health, most researchers have focused on specific bacterial taxa or on a single body habitat, the gut. Thus, the initiation stage of human microbiome development remains obscure. The goal of the present study was to obtain a community-wide perspective on the influence of delivery mode and body habitat on the neonate's first microbiota. We used multiplexed 16S rRNA gene pyrosequencing to characterize bacterial communities from mothers and their newborn babies, four born vaginally and six born via Cesarean section. Mothers' skin, oral mucosa, and vagina were sampled 1 h before delivery, and neonates' skin, oral mucosa, and nasopharyngeal aspirate were sampled {$<$}5 min, and meconium {$<$}24 h, after delivery. We found that in direct contrast to the highly differentiated communities of their mothers, neonates harbored bacterial communities that were undifferentiated across multiple body habitats, regardless of delivery mode. Our results also show that vaginally delivered infants acquired bacterial communities resembling their own mother's vaginal microbiota, dominated by Lactobacillus, Prevotella, or Sneathia spp., and C-section infants harbored bacterial communities similar to those found on the skin surface, dominated by Staphylococcus, Corynebacterium, and Propionibacterium spp. These findings establish an important baseline for studies tracking the human microbiome's successional development in different body habitats following different delivery modes, and their associated effects on infant health.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
keywords = {菌群发育,里程碑},
language = {eng},
number = {26},
pmcid = {PMC2900693},
pmid = {20566857}
}
@article{dominguez-belloPartialRestorationMicrobiota2016,
title = {Partial Restoration of the Microbiota of Cesarean-Born Infants via Vaginal Microbial Transfer},
author = {{Dominguez-Bello}, Maria G. and {De Jesus-Laboy}, Kassandra M. and Shen, Nan and Cox, Laura M. and Amir, Amnon and Gonzalez, Antonio and Bokulich, Nicholas A. and Song, Se Jin and Hoashi, Marina and {Rivera-Vinas}, Juana I. and Mendez, Keimari and Knight, Rob and Clemente, Jose C.},
year = {2016},
month = mar,
volume = {22},
pages = {250--253},
issn = {1546-170X},
doi = {10.1038/nm.4039},
abstract = {Exposure of newborns to the maternal vaginal microbiota is interrupted with cesarean birthing. Babies delivered by cesarean section (C-section) acquire a microbiota that differs from that of vaginally delivered infants, and C-section delivery has been associated with increased risk for immune and metabolic disorders. Here we conducted a pilot study in which infants delivered by C-section were exposed to maternal vaginal fluids at birth. Similarly to vaginally delivered babies, the gut, oral and skin bacterial communities of these newborns during the first 30 d of life was enriched in vaginal bacteria--which were underrepresented in unexposed C-section-delivered infants--and the microbiome similarity to those of vaginally delivered infants was greater in oral and skin samples than in anal samples. Although the long-term health consequences of restoring the microbiota of C-section-delivered infants remain unclear, our results demonstrate that vaginal microbes can be partially restored at birth in C-section-delivered babies.},
journal = {Nature Medicine},
keywords = {菌群发育,里程碑},
language = {eng},
number = {3},
pmcid = {PMC5062956},
pmid = {26828196}
}
@article{durgandavidj.RoleGutMicrobiome2016,
title = {Role of the {{Gut Microbiome}} in {{Obstructive Sleep Apnea}}\textendash{{Induced Hypertension}}},
author = {{Durgan David J.} and {Ganesh Bhanu P.} and {Cope Julia L.} and {Ajami Nadim J.} and {Phillips Sharon C.} and {Petrosino Joseph F.} and {Hollister Emily B.} and {Bryan Robert M.}},
year = {2016},
month = feb,
volume = {67},
pages = {469--474},
publisher = {{American Heart Association}},
doi = {10.1161/HYPERTENSIONAHA.115.06672},
abstract = {Individuals suffering from obstructive sleep apnea (OSA) are at increased risk for systemic hypertension. The importance of a healthy gut microbiota, and detriment of a dysbiotic microbiota, on host physiology is becoming increasingly evident. We tested the hypothesis that gut dysbiosis contributes to hypertension observed with OSA. OSA was modeled in rats by inflating a tracheal balloon during the sleep cycle (10-s inflations, 60 per hour). On normal chow diet, OSA had no effect on blood pressure; however, in rats fed a high-fat diet, blood pressure increased 24 and 29 mm Hg after 7 and 14 days of OSA, respectively (P{$<$}0.05 each). Bacterial community characterization was performed on fecal pellets isolated before and after 14 days of OSA in chow and high-fat fed rats. High-fat diet and OSA led to significant alterations of the gut microbiota, including decreases in bacterial taxa known to produce the short chain fatty acid butyrate (P{$<$}0.05). Finally, transplant of dysbiotic cecal contents from hypertensive OSA rats on high-fat diet into OSA recipient rats on normal chow diet (shown to be normotensive) resulted in hypertension similar to that of the donor (increased 14 and 32 mm Hg after 7 and 14 days of OSA, respectively; P{$<$}0.05). These studies demonstrate a causal relationship between gut dysbiosis and hypertension, and suggest that manipulation of the microbiota may be a viable treatment for OSA-induced, and possibly other forms of, hypertension.},
journal = {Hypertension},
number = {2}
}
@article{earleQuantitativeImagingGut2015,
title = {Quantitative {{Imaging}} of {{Gut Microbiota Spatial Organization}}},
author = {Earle, Kristen A. and Billings, Gabriel and Sigal, Michael and Lichtman, Joshua S. and Hansson, Gunnar C. and Elias, Joshua E. and Amieva, Manuel R. and Huang, Kerwyn Casey and Sonnenburg, Justin L.},
year = {2015},
month = oct,
volume = {18},
pages = {478--488},
issn = {19313128},
doi = {10.1016/j.chom.2015.09.002},
journal = {Cell Host \& Microbe},
language = {en},
number = {4}
}
@article{eckburgDiversityHumanIntestinal2005,
title = {Diversity of the {{Human Intestinal Microbial Flora}}},
author = {Eckburg, Paul B. and Bik, Elisabeth M. and Bernstein, Charles N. and Purdom, Elizabeth and Dethlefsen, Les and Sargent, Michael and Gill, Steven R. and Nelson, Karen E. and Relman, David A.},
year = {2005},
month = jun,
volume = {308},
pages = {1635--1638},
issn = {0036-8075},
doi = {10.1126/science.1110591},
abstract = {The human endogenous intestinal microflora is an essential ``organ'' in providing nourishment, regulating epithelial development, and instructing innate immunity; yet, surprisingly, basic features remain poorly described. We examined 13,355 prokaryotic ribosomal RNA gene sequences from multiple colonic mucosal sites and feces of healthy subjects to improve our understanding of gut microbial diversity. A majority of the bacterial sequences corresponded to uncultivated species and novel microorganisms. We discovered significant intersubject variability and differences between stool and mucosa community composition. Characterization of this immensely diverse ecosystem is the first step in elucidating its role in health and disease.},
journal = {Science (New York, N.Y.)},
keywords = {菌群稳定性,里程碑},
number = {5728},
pmcid = {PMC1395357},
pmid = {15831718}
}
@article{everardCrosstalkAkkermansiaMuciniphila2013,
title = {Cross-Talk between {{Akkermansia}} Muciniphila and Intestinal Epithelium Controls Diet-Induced Obesity},
author = {Everard, Amandine and Belzer, Clara and Geurts, Lucie and Ouwerkerk, Janneke P. and Druart, C{\'e}line and Bindels, Laure B. and Guiot, Yves and Derrien, Muriel and Muccioli, Giulio G. and Delzenne, Nathalie M. and de Vos, Willem M. and Cani, Patrice D.},
year = {2013},
month = may,
volume = {110},
pages = {9066--9071},
publisher = {{National Academy of Sciences}},
issn = {0027-8424, 1091-6490},
doi = {10.1073/pnas.1219451110},
abstract = {Obesity and type 2 diabetes are characterized by altered gut microbiota, inflammation, and gut barrier disruption. Microbial composition and the mechanisms of interaction with the host that affect gut barrier function during obesity and type 2 diabetes have not been elucidated. We recently isolated Akkermansia muciniphila, which is a mucin-degrading bacterium that resides in the mucus layer. The presence of this bacterium inversely correlates with body weight in rodents and humans. However, the precise physiological roles played by this bacterium during obesity and metabolic disorders are unknown. This study demonstrated that the abundance of A. muciniphila decreased in obese and type 2 diabetic mice. We also observed that prebiotic feeding normalized A. muciniphila abundance, which correlated with an improved metabolic profile. In addition, we demonstrated that A. muciniphila treatment reversed high-fat diet-induced metabolic disorders, including fat-mass gain, metabolic endotoxemia, adipose tissue inflammation, and insulin resistance. A. muciniphila administration increased the intestinal levels of endocannabinoids that control inflammation, the gut barrier, and gut peptide secretion. Finally, we demonstrated that all these effects required viable A. muciniphila because treatment with heat-killed cells did not improve the metabolic profile or the mucus layer thickness. In summary, this study provides substantial insight into the intricate mechanisms of bacterial (i.e., A. muciniphila) regulation of the cross-talk between the host and gut microbiota. These results also provide a rationale for the development of a treatment that uses this human mucus colonizer for the prevention or treatment of obesity and its associated metabolic disorders.},
chapter = {Biological Sciences},
copyright = {\textcopyright{} . Freely available online through the PNAS open access option.},
journal = {Proceedings of the National Academy of Sciences},
language = {en},
number = {22},
pmid = {23671105}
}
@article{everardResponsesGutMicrobiota2011,
title = {Responses of {{Gut Microbiota}} and {{Glucose}} and {{Lipid Metabolism}} to {{Prebiotics}} in {{Genetic Obese}} and {{Diet}}-{{Induced Leptin}}-{{Resistant Mice}}},
author = {Everard, Amandine and Lazarevic, Vladimir and Derrien, Muriel and Girard, Myriam and Muccioli, Giulio G. and Neyrinck, Audrey M. and Possemiers, Sam and Holle, Ann Van and Fran{\c c}ois, Patrice and de Vos, Willem M. and Delzenne, Nathalie M. and Schrenzel, Jacques and Cani, Patrice D.},
year = {2011},
month = nov,
volume = {60},
pages = {2775--2786},
publisher = {{American Diabetes Association}},
issn = {0012-1797, 1939-327X},
doi = {10.2337/db11-0227},
abstract = {OBJECTIVE To investigate deep and comprehensive analysis of gut microbial communities and biological parameters after prebiotic administration in obese and diabetic mice. RESEARCH DESIGN AND METHODS Genetic (ob/ob) or diet-induced obese and diabetic mice were chronically fed with prebiotic-enriched diet or with a control diet. Extensive gut microbiota analyses, including quantitative PCR, pyrosequencing of the 16S rRNA, and phylogenetic microarrays, were performed in ob/ob mice. The impact of gut microbiota modulation on leptin sensitivity was investigated in diet-induced leptin-resistant mice. Metabolic parameters, gene expression, glucose homeostasis, and enteroendocrine-related L-cell function were documented in both models. RESULTS In ob/ob mice, prebiotic feeding decreased Firmicutes and increased Bacteroidetes phyla, but also changed 102 distinct taxa, 16 of which displayed a {$>$}10-fold change in abundance. In addition, prebiotics improved glucose tolerance, increased L-cell number and associated parameters (intestinal proglucagon mRNA expression and plasma glucagon-like peptide-1 levels), and reduced fat-mass development, oxidative stress, and low-grade inflammation. In high fat\textendash fed mice, prebiotic treatment improved leptin sensitivity as well as metabolic parameters. CONCLUSIONS We conclude that specific gut microbiota modulation improves glucose homeostasis, leptin sensitivity, and target enteroendocrine cell activity in obese and diabetic mice. By profiling the gut microbiota, we identified a catalog of putative bacterial targets that may affect host metabolism in obesity and diabetes.},
chapter = {Metabolism},
copyright = {\textcopyright{} 2011 by the American Diabetes Association.. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.},
journal = {Diabetes},
language = {en},
number = {11},
pmid = {21933985}
}
@article{faithLongtermStabilityHuman2013,
title = {The Long-Term Stability of the Human Gut Microbiota},
author = {Faith, Jeremiah J. and Guruge, Janaki L. and Charbonneau, Mark and Subramanian, Sathish and Seedorf, Henning and Goodman, Andrew L. and Clemente, Jose C. and Knight, Rob and Heath, Andrew C. and Leibel, Rudolph L. and Rosenbaum, Michael and Gordon, Jeffrey I.},
year = {2013},
month = jul,
volume = {341},
pages = {1237439},
issn = {1095-9203},
doi = {10.1126/science.1237439},
abstract = {A low-error 16S ribosomal RNA amplicon sequencing method, in combination with whole-genome sequencing of {$>$}500 cultured isolates, was used to characterize bacterial strain composition in the fecal microbiota of 37 U.S. adults sampled for up to 5 years. Microbiota stability followed a power-law function, which when extrapolated suggests that most strains in an individual are residents for decades. Shared strains were recovered from family members but not from unrelated individuals. Sampling of individuals who consumed a monotonous liquid diet for up to 32 weeks indicated that changes in strain composition were better predicted by changes in weight than by differences in sampling interval. This combination of stability and responsiveness to physiologic change confirms the potential of the gut microbiota as a diagnostic tool and therapeutic target.},
journal = {Science (New York, N.Y.)},
keywords = {菌群稳定性,里程碑},
language = {eng},
number = {6141},
pmcid = {PMC3791589},
pmid = {23828941}
}
@article{falonyPopulationlevelAnalysisGut2016,
title = {Population-Level Analysis of Gut Microbiome Variation},
author = {Falony, G. and Joossens, M. and {Vieira-Silva}, S. and Wang, J. and Darzi, Y. and Faust, K. and Kurilshikov, A. and Bonder, M. J. and {Valles-Colomer}, M. and Vandeputte, D. and Tito, R. Y. and Chaffron, S. and Rymenans, L. and Verspecht, C. and De Sutter, L. and {Lima-Mendez}, G. and Dhoe, K. and Jonckheere, K. and Homola, D. and Garcia, R. and Tigchelaar, E. F. and Eeckhaudt, L. and Fu, J. and Henckaerts, L. and Zhernakova, A. and Wijmenga, C. and Raes, J.},
year = {2016},
month = apr,
volume = {352},
pages = {560--564},
issn = {0036-8075, 1095-9203},
doi = {10.1126/science.aad3503},
abstract = {``Normal'' for the gut microbiota For the benefit of future clinical studies, it is critical to establish what constitutes a ``normal'' gut microbiome, if it exists at all. Through fecal samples and questionnaires, Falony et al. and Zhernakova et al. targeted general populations in Belgium and the Netherlands, respectively. Gut microbiota composition correlated with a range of factors including diet, use of medication, red blood cell counts, fecal chromogranin A, and stool consistency. The data give some hints for possible biomarkers of normal gut communities. Science, this issue pp. 560 and 565 Fecal microbiome variation in the average, healthy population has remained under-investigated. Here, we analyzed two independent, extensively phenotyped cohorts: the Belgian Flemish Gut Flora Project (FGFP; discovery cohort; N = 1106) and the Dutch LifeLines-DEEP study (LLDeep; replication; N = 1135). Integration with global data sets (N combined = 3948) revealed a 14-genera core microbiota, but the 664 identified genera still underexplore total gut diversity. Sixty-nine clinical and questionnaire-based covariates were found associated to microbiota compositional variation with a 92\% replication rate. Stool consistency showed the largest effect size, whereas medication explained largest total variance and interacted with other covariate-microbiota associations. Early-life events such as birth mode were not reflected in adult microbiota composition. Finally, we found that proposed disease marker genera associated to host covariates, urging inclusion of the latter in study design. Two large-scale studies in Western Europe establish environment-diet-microbe-host interactions. Two large-scale studies in Western Europe establish environment-diet-microbe-host interactions.},
file = {E\:\\zotero.storage.2016\\Science\\2016\\2016_Population-level analysis of gut microbiome variation.pdf},
journal = {Science},
language = {en},
number = {6285}
}
@article{farrellImportanceFeedingYour2019,
title = {The Importance of Feeding Your Microbiota},
author = {Farrell, Alison},
year = {2019},
month = jun,
publisher = {{Nature Publishing Group}},
doi = {10.1038/d42859-019-00015-1},
abstract = {Discover the world's best science and medicine | Nature.com},
copyright = {2019 Nature},
journal = {Nature Research},
keywords = {喂养菌群,里程碑},
language = {en}
}
@article{ferrettiMothertoInfantMicrobialTransmission2018,
title = {Mother-to-{{Infant Microbial Transmission}} from {{Different Body Sites Shapes}} the {{Developing Infant Gut Microbiome}}},
author = {Ferretti, Pamela and Pasolli, Edoardo and Tett, Adrian and Asnicar, Francesco and Gorfer, Valentina and Fedi, Sabina and Armanini, Federica and Truong, Duy Tin and Manara, Serena and Zolfo, Moreno and Beghini, Francesco and Bertorelli, Roberto and De Sanctis, Veronica and Bariletti, Ilaria and Canto, Rosarita and Clementi, Rosanna and Cologna, Marina and Crif{\`o}, Tiziana and Cusumano, Giuseppina and Gottardi, Stefania and Innamorati, Claudia and Mas{\`e}, Caterina and Postai, Daniela and Savoi, Daniela and Duranti, Sabrina and Lugli, Gabriele Andrea and Mancabelli, Leonardo and Turroni, Francesca and Ferrario, Chiara and Milani, Christian and Mangifesta, Marta and Anzalone, Rosaria and Viappiani, Alice and Yassour, Moran and Vlamakis, Hera and Xavier, Ramnik and Collado, Carmen Maria and Koren, Omry and Tateo, Saverio and Soffiati, Massimo and Pedrotti, Anna and Ventura, Marco and Huttenhower, Curtis and Bork, Peer and Segata, Nicola},
year = {2018},
month = nov,
volume = {24},
pages = {133-145.e5},
issn = {1934-6069},
doi = {10.1016/j.chom.2018.06.005},
abstract = {The acquisition and development of the infant microbiome are key to establishing a healthy host-microbiome symbiosis. The maternal microbial reservoir is thought to play a crucial role in this process. However, the source and transmission routes of the infant pioneering microbes are poorly understood. To address this, we longitudinally sampled the microbiome of 25 mother-infant pairs~across multiple body sites from birth up to 4~months postpartum. Strain-level metagenomic profiling showed a rapid influx of microbes at birth~followed by strong selection during the first few days of life. Maternal skin and vaginal strains~colonize only transiently, and the infant continues to acquire microbes from distinct maternal sources after birth. Maternal gut strains proved more persistent in the infant gut and ecologically better adapted than those acquired from other sources. Together, these data describe the mother-to-infant microbiome transmission routes that are integral in the development of the infant microbiome.},
journal = {Cell Host \& Microbe},
keywords = {菌群发育,里程碑},
language = {eng},
number = {1},
pmcid = {PMC6716579},
pmid = {30001516}
}
@article{filippoImpactDietShaping2010,
title = {Impact of Diet in Shaping Gut Microbiota Revealed by a Comparative Study in Children from {{Europe}} and Rural {{Africa}}},
author = {Filippo, Carlotta De and Cavalieri, Duccio and Paola, Monica Di and Ramazzotti, Matteo and Poullet, Jean Baptiste and Massart, Sebastien and Collini, Silvia and Pieraccini, Giuseppe and Lionetti, Paolo},
year = {2010},
month = aug,
volume = {107},
pages = {14691--14696},
publisher = {{National Academy of Sciences}},
issn = {0027-8424, 1091-6490},
doi = {10.1073/pnas.1005963107},
abstract = {Gut microbial composition depends on different dietary habits just as health depends on microbial metabolism, but the association of microbiota with different diets in human populations has not yet been shown. In this work, we compared the fecal microbiota of European children (EU) and that of children from a rural African village of Burkina Faso (BF), where the diet, high in fiber content, is similar to that of early human settlements at the time of the birth of agriculture. By using high-throughput 16S rDNA sequencing and biochemical analyses, we found significant differences in gut microbiota between the two groups. BF children showed a significant enrichment in Bacteroidetes and depletion in Firmicutes (P {$<$} 0.001), with a unique abundance of bacteria from the genus Prevotella and Xylanibacter, known to contain a set of bacterial genes for cellulose and xylan hydrolysis, completely lacking in the EU children. In addition, we found significantly more short-chain fatty acids (P {$<$} 0.001) in BF than in EU children. Also, Enterobacteriaceae (Shigella and Escherichia) were significantly underrepresented in BF than in EU children (P {$<$} 0.05). We hypothesize that gut microbiota coevolved with the polysaccharide-rich diet of BF individuals, allowing them to maximize energy intake from fibers while also protecting them from inflammations and noninfectious colonic diseases. This study investigates and compares human intestinal microbiota from children characterized by a modern western diet and a rural diet, indicating the importance of preserving this treasure of microbial diversity from ancient rural communities worldwide.},
chapter = {Biological Sciences},
copyright = {\textcopyright{} . Freely available online through the PNAS open access option.},
journal = {Proceedings of the National Academy of Sciences},
language = {en},
number = {33},
pmid = {20679230}
}
@article{forslundDisentanglingTypeDiabetes2015a,
title = {Disentangling Type 2 Diabetes and Metformin Treatment Signatures in the Human Gut Microbiota},
author = {Forslund, Kristoffer and Hildebrand, Falk and Nielsen, Trine and Falony, Gwen and Le Chatelier, Emmanuelle and Sunagawa, Shinichi and Prifti, Edi and {Vieira-Silva}, Sara and Gudmundsdottir, Valborg and Krogh Pedersen, Helle and Arumugam, Manimozhiyan and Kristiansen, Karsten and Yvonne Voigt, Anita and Vestergaard, Henrik and Hercog, Rajna and Igor Costea, Paul and Roat Kultima, Jens and Li, Junhua and J{\o}rgensen, Torben and Levenez, Florence and Dore, Jo{\"e}l and Bj{\o}rn Nielsen, H. and Brunak, S{\o}ren and Raes, Jeroen and Hansen, Torben and Wang, Jun and Dusko Ehrlich, S. and Bork, Peer and Pedersen, Oluf},
year = {2015},
month = dec,
volume = {528},
pages = {262--266},
publisher = {{Nature Publishing Group}},
issn = {1476-4687},
doi = {10.1038/nature15766},
abstract = {Growing evidence from metagenome-wide association studies link multiple common disorders to microbial dysbiosis but effects of drug treatment are often not accounted for; here, the authors re-analyse two previous metagenomic studies of type 2 diabetes mellitus patients together with a novel cohort to determine the effects of the widely prescribed antidiabetic drug metformin and highlight the need to distinguish the effects of a disease from the effects of treatment on the gut microbiota.},
copyright = {2015 Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved.},
journal = {Nature},
language = {en},
number = {7581}
}
@article{frankMolecularphylogeneticCharacterizationMicrobial2007,
title = {Molecular-Phylogenetic Characterization of Microbial Community Imbalances in Human Inflammatory Bowel Diseases},
author = {Frank, Daniel N. and Amand, Allison L. St and Feldman, Robert A. and Boedeker, Edgar C. and Harpaz, Noam and Pace, Norman R.},
year = {2007},
month = aug,
volume = {104},
pages = {13780--13785},
publisher = {{National Academy of Sciences}},
issn = {0027-8424, 1091-6490},
doi = {10.1073/pnas.0706625104},
abstract = {The two primary human inflammatory bowel diseases, Crohn's disease (CD) and ulcerative colitis (UC), are idiopathic relapsing disorders characterized by chronic inflammation of the intestinal tract. Although several lines of reasoning suggest that gastrointestinal (GI) microbes influence inflammatory bowel disease (IBD) pathogenesis, the types of microbes involved have not been adequately described. Here we report the results of a culture-independent rRNA sequence analysis of GI tissue samples obtained from CD and UC patients, as well as non-IBD controls. Specimens were obtained through surgery from a variety of intestinal sites and included both pathologically normal and abnormal states. Our results provide comprehensive molecular-based analysis of the microbiota of the human small intestine. Comparison of clone libraries reveals statistically significant differences between the microbiotas of CD and UC patients and those of non-IBD controls. Significantly, our results indicate that a subset of CD and UC samples contained abnormal GI microbiotas, characterized by depletion of commensal bacteria, notably members of the phyla Firmicutes and Bacteroidetes. Patient stratification by GI microbiota provides further evidence that CD represents a spectrum of disease states and suggests that treatment of some forms of IBD may be facilitated by redress of the detected microbiological imbalances.},
chapter = {Biological Sciences},
copyright = {\textcopyright{} 2007 by The National Academy of Sciences of the USA},
journal = {Proceedings of the National Academy of Sciences},
language = {en},
number = {34},
pmid = {17699621}
}
@article{fredricksMolecularIdentificationBacteria2005,
title = {Molecular Identification of Bacteria Associated with Bacterial Vaginosis},
author = {Fredricks, David N. and Fiedler, Tina L. and Marrazzo, Jeanne M.},
year = {2005},
month = nov,
volume = {353},
pages = {1899--1911},
issn = {1533-4406},
doi = {10.1056/NEJMoa043802},
abstract = {BACKGROUND: Bacterial vaginosis affects millions of women and is associated with several serious health conditions. The cause of bacterial vaginosis remains poorly understood despite numerous studies based on cultures. Bacteria in microbial communities can be identified without cultivation by characterizing their ribosomal DNA (rDNA) sequences. METHODS: We identified bacteria in samples of vaginal fluid with a combination of broad-range polymerase-chain-reaction (PCR) amplification of 16S rDNA with clone analysis, bacterium-specific PCR assay of 16S rDNA, and fluorescence in situ hybridization (FISH) performed directly on vaginal fluid from 27 subjects with bacterial vaginosis and 46 without the condition. Twenty-one subjects were studied with the use of broad-range PCR of 16S rDNA, and 73 subjects were studied with the use of bacterium-specific PCR. RESULTS: Women without bacterial vaginosis had 1 to 6 vaginal bacterial species (phylotypes) in each sample (mean, 3.3), as detected by broad-range PCR of 16S rDNA, and lactobacillus species were the predominant bacteria noted (83 to 100 percent of clones). Women with bacterial vaginosis had greater bacterial diversity (P{$<$}0.001), with 9 to 17 phylotypes (mean, 12.6) detected per sample and newly recognized species present in 32 to 89 percent of clones per sample library (mean, 58 percent). Thirty-five unique bacterial species were detected in the women with bacterial vaginosis, including several species with no close cultivated relatives. Bacterium-specific PCR assays showed that several bacteria that had not been previously described were highly prevalent in subjects with bacterial vaginosis but rare in healthy controls. FISH confirmed that newly recognized bacteria detected by PCR corresponded to specific bacterial morphotypes visible in vaginal fluid. CONCLUSIONS: Women with bacterial vaginosis have complex vaginal infections with many newly recognized species, including three bacteria in the Clostridiales order that were highly specific for bacterial vaginosis.},
journal = {The New England Journal of Medicine},
keywords = {技术进步,里程碑},
language = {eng},
number = {18},
pmid = {16267321}
}
@article{fujimuraMicrobiotaAllergyAsthma2015,
title = {Microbiota in {{Allergy}} and {{Asthma}} and the {{Emerging Relationship}} with the {{Gut Microbiome}}},
author = {Fujimura, Kei E. and Lynch, Susan V.},
year = {2015},
month = may,
volume = {17},
pages = {592--602},
issn = {19313128},
doi = {10.1016/j.chom.2015.04.007},
journal = {Cell Host \& Microbe},
language = {en},
number = {5}
}
@article{furetDifferentialAdaptationHuman2010,
title = {Differential {{Adaptation}} of {{Human Gut Microbiota}} to {{Bariatric Surgery}}\textendash{{Induced Weight Loss}}: {{Links With Metabolic}} and {{Low}}-{{Grade Inflammation Markers}}},
shorttitle = {Differential {{Adaptation}} of {{Human Gut Microbiota}} to {{Bariatric Surgery}}\textendash{{Induced Weight Loss}}},
author = {Furet, Jean-Pierre and Kong, Ling-Chun and Tap, Julien and Poitou, Christine and Basdevant, Arnaud and Bouillot, Jean-Luc and Mariat, Denis and Corthier, G{\'e}rard and Dor{\'e}, Jo{\"e}l and Henegar, Corneliu and Rizkalla, Salwa and Cl{\'e}ment, Karine},
year = {2010},
month = dec,
volume = {59},
pages = {3049--3057},
publisher = {{American Diabetes Association}},
issn = {0012-1797, 1939-327X},
doi = {10.2337/db10-0253},
abstract = {OBJECTIVE Obesity alters gut microbiota ecology and associates with low-grade inflammation in humans. Roux-en-Y gastric bypass (RYGB) surgery is one of the most efficient procedures for the treatment of morbid obesity resulting in drastic weight loss and improvement of metabolic and inflammatory status. We analyzed the impact of RYGB on the modifications of gut microbiota and examined links with adaptations associated with this procedure. RESEARCH DESIGN AND METHODS Gut microbiota was profiled from fecal samples by real-time quantitative PCR in 13 lean control subjects and in 30 obese individuals (with seven type 2 diabetics) explored before (M0), 3 months (M3), and 6 months (M6) after RYGB. RESULTS Four major findings are highlighted: 1) Bacteroides/Prevotella group was lower in obese subjects than in control subjects at M0 and increased at M3. It was negatively correlated with corpulence, but the correlation depended highly on caloric intake; 2) Escherichia coli species increased at M3 and inversely correlated with fat mass and leptin levels independently of changes in food intake; 3) lactic acid bacteria including Lactobacillus/Leuconostoc/Pediococcus group and Bifidobacterium genus decreased at M3; and 4) Faecalibacterium prausnitzii species was lower in subjects with diabetes and associated negatively with inflammatory markers at M0 and throughout the follow-up after surgery independently of changes in food intake. CONCLUSIONS These results suggest that components of the dominant gut microbiota rapidly adapt in a starvation-like situation induced by RYGB while the F. prausnitzii species is directly linked to the reduction in low-grade inflammation state in obesity and diabetes independently of calorie intake.},
chapter = {Obesity Studies},
copyright = {\textcopyright{} 2010 by the American Diabetes Association.. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.},
journal = {Diabetes},
language = {en},
number = {12},
pmid = {20876719}
}
@article{gajerTemporalDynamicsHuman2012,
title = {Temporal Dynamics of the Human Vaginal Microbiota},
author = {Gajer, Pawel and Brotman, Rebecca M. and Bai, Guoyun and Sakamoto, Joyce and Sch{\"u}tte, Ursel M. E. and Zhong, Xue and Koenig, Sara S. K. and Fu, Li and Ma, Zhanshan Sam and Zhou, Xia and Abdo, Zaid and Forney, Larry J. and Ravel, Jacques},
year = {2012},
month = may,
volume = {4},
pages = {132ra52},
issn = {1946-6242},
doi = {10.1126/scitranslmed.3003605},
abstract = {Elucidating the factors that impinge on the stability of bacterial communities in the vagina may help in predicting the risk of diseases that affect women's health. Here, we describe the temporal dynamics of the composition of vaginal bacterial communities in 32 reproductive-age women over a 16-week period. The analysis revealed the dynamics of five major classes of bacterial communities and showed that some communities change markedly over short time periods, whereas others are relatively stable. Modeling community stability using new quantitative measures indicates that deviation from stability correlates with time in the menstrual cycle, bacterial community composition, and sexual activity. The women studied are healthy; thus, it appears that neither variation in community composition per se nor higher levels of observed diversity (co-dominance) are necessarily indicative of dysbiosis.},
journal = {Science Translational Medicine},
keywords = {菌群稳定性,里程碑},
language = {eng},
number = {132},
pmcid = {PMC3722878},
pmid = {22553250}
}
@article{gaoMolecularAnalysisHuman2007,
title = {Molecular Analysis of Human Forearm Superficial Skin Bacterial Biota},
author = {Gao, Zhan and Tseng, Chi-hong and Pei, Zhiheng and Blaser, Martin J.},
year = {2007},
month = feb,
volume = {104},
pages = {2927--2932},
issn = {0027-8424},
doi = {10.1073/pnas.0607077104},
abstract = {The microbial ecology of human skin is complex, but little is known about its species composition. We examined the diversity of the skin biota from the superficial volar left and right forearms in six healthy subjects using broad-range small subunit rRNA genes (16S rDNA) PCR-based sequencing of randomly selected clones. For the initial 1,221 clones analyzed, 182 species-level operational taxonomic units (SLOTUs) belonging to eight phyla were identified, estimated as 74.0\% [95\% confidence interval (C.I.), approximately 64.8-77.9\%] of the SLOTUs in this ecosystem; 48.0 +/- 12.2 SLOTUs were found in each subject. Three phyla (Actinobacteria, Firmicutes, and Proteobacteria) accounted for 94.6\% of the clones. Most (85.3\%) of the bacterial sequences corresponded to known and cultivated species, but 98 (8.0\%) clones, comprising 30 phylotypes, had {$<$}97\% similarity to prior database sequences. Only 6 (6.6\%) of the 91 genera and 4 (2.2\%) of the 182 SLOTUs, respectively, were found in all six subjects. Analysis of 817 clones obtained 8-10 months later from four subjects showed additional phyla (numbering 2), genera (numbering 28), and SLOTUs (numbering 65). Only four (3.4\%) of the 119 genera (Propionibacteria, Corynebacteria, Staphylococcus, and Streptococcus) were observed in each subject tested twice, but these genera represented 54.4\% of all clones. These results show that the bacterial biota in normal superficial skin is highly diverse, with few well conserved and well represented genera, but otherwise low-level interpersonal consensus.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
keywords = {技术进步,里程碑},
language = {eng},
number = {8},
pmcid = {PMC1815283},
pmid = {17293459}
}
@article{gensollenHowColonizationMicrobiota2016,
title = {How Colonization by Microbiota in Early Life Shapes the Immune System},
author = {Gensollen, T. and Iyer, S. S. and Kasper, D. L. and Blumberg, R. S.},
year = {2016},
month = apr,
volume = {352},
pages = {539--544},
issn = {0036-8075, 1095-9203},
doi = {10.1126/science.aad9378},
journal = {Science},
language = {en},
number = {6285}
}
@article{geversTreatmentNaiveMicrobiomeNewOnset2014,
title = {The {{Treatment}}-{{Naive Microbiome}} in {{New}}-{{Onset Crohn}}'s {{Disease}}},
author = {Gevers, Dirk and Kugathasan, Subra and Denson, Lee A. and {V{\'a}zquez-Baeza}, Yoshiki and Van Treuren, Will and Ren, Boyu and Schwager, Emma and Knights, Dan and Song, Se Jin and Yassour, Moran and Morgan, Xochitl C. and Kostic, Aleksandar D. and Luo, Chengwei and Gonz{\'a}lez, Antonio and McDonald, Daniel and Haberman, Yael and Walters, Thomas and Baker, Susan and Rosh, Joel and Stephens, Michael and Heyman, Melvin and Markowitz, James and Baldassano, Robert and Griffiths, Anne and Sylvester, Francisco and Mack, David and Kim, Sandra and Crandall, Wallace and Hyams, Jeffrey and Huttenhower, Curtis and Knight, Rob and Xavier, Ramnik J.},
year = {2014},
month = mar,
volume = {15},
pages = {382--392},
publisher = {{Elsevier}},
issn = {1931-3128},
doi = {10.1016/j.chom.2014.02.005},
journal = {Cell Host \& Microbe},
language = {English},
number = {3},
pmid = {24629344}
}
@article{ghannoumCharacterizationOralFungal2010,
title = {Characterization of the Oral Fungal Microbiome (Mycobiome) in Healthy Individuals},
author = {Ghannoum, Mahmoud A. and Jurevic, Richard J. and Mukherjee, Pranab K. and Cui, Fan and Sikaroodi, Masoumeh and Naqvi, Ammar and Gillevet, Patrick M.},
year = {2010},
month = jan,
volume = {6},
pages = {e1000713},
issn = {1553-7374},
doi = {10.1371/journal.ppat.1000713},
abstract = {The oral microbiome-organisms residing in the oral cavity and their collective genome-are critical components of health and disease. The fungal component of the oral microbiota has not been characterized. In this study, we used a novel multitag pyrosequencing approach to characterize fungi present in the oral cavity of 20 healthy individuals, using the pan-fungal internal transcribed spacer (ITS) primers. Our results revealed the "basal" oral mycobiome profile of the enrolled individuals, and showed that across all the samples studied, the oral cavity contained 74 culturable and 11 non-culturable fungal genera. Among these genera, 39 were present in only one person, 16 genera were present in two participants, and 5 genera were present in three people, while 15 genera (including non-culturable organisms) were present in {$>$}/=4 (20\%) participants. Candida species were the most frequent (isolated from 75\% of participants), followed by Cladosporium (65\%), Aureobasidium, Saccharomycetales (50\% for both), Aspergillus (35\%), Fusarium (30\%), and Cryptococcus (20\%). Four of these predominant genera are known to be pathogenic in humans. The low-abundance genera may represent environmental fungi present in the oral cavity and could simply be spores inhaled from the air or material ingested with food. Among the culturable genera, 61 were represented by one species each, while 13 genera comprised between 2 and 6 different species; the total number of species identified were 101. The number of species in the oral cavity of each individual ranged between 9 and 23. Principal component (PCO) analysis of the obtained data set followed by sample clustering and UniFrac analysis revealed that White males and Asian males clustered differently from each other, whereas both Asian and White females clustered together. This is the first study that identified the "basal mycobiome" of healthy individuals, and provides the basis for a detailed characterization of the oral mycobiome in health and disease.},
journal = {PLoS pathogens},
keywords = {真菌和病毒,里程碑},
language = {eng},
number = {1},
pmcid = {PMC2795202},
pmid = {20072605}
}
@article{giongoDefiningAutoimmuneMicrobiome2011,
title = {Toward Defining the Autoimmune Microbiome for Type 1 Diabetes},
author = {Giongo, Adriana and Gano, Kelsey A. and Crabb, David B. and Mukherjee, Nabanita and Novelo, Luis L. and Casella, George and Drew, Jennifer C. and Ilonen, Jorma and Knip, Mikael and Hy{\"o}ty, Heikki and Veijola, Riitta and Simell, Tuula and Simell, Olli and Neu, Josef and Wasserfall, Clive H. and Schatz, Desmond and Atkinson, Mark A. and Triplett, Eric W.},
year = {2011},
month = jan,
volume = {5},
pages = {82--91},
publisher = {{Nature Publishing Group}},
issn = {1751-7370},
doi = {10.1038/ismej.2010.92},
abstract = {Several studies have shown that gut bacteria have a role in diabetes in murine models. Specific bacteria have been correlated with the onset of diabetes in a rat model. However, it is unknown whether human intestinal microbes have a role in the development of autoimmunity that often leads to type 1 diabetes (T1D), an autoimmune disorder in which insulin-secreting pancreatic islet cells are destroyed. High-throughput, culture-independent approaches identified bacteria that correlate with the development of T1D-associated autoimmunity in young children who are at high genetic risk for this disorder. The level of bacterial diversity diminishes overtime in these autoimmune subjects relative to that of age-matched, genotype-matched, nonautoimmune individuals. A single species, Bacteroides ovatus, comprised nearly 24\% of the total increase in the phylum Bacteroidetes in cases compared with controls. Conversely, another species in controls, represented by the human firmicute strain CO19, represented nearly 20\% of the increase in Firmicutes compared with cases overtime. Three lines of evidence are presented that support the notion that, as healthy infants approach the toddler stage, their microbiomes become healthier and more stable, whereas, children who are destined for autoimmunity develop a microbiome that is less diverse and stable. Hence, the autoimmune microbiome for T1D may be distinctly different from that found in healthy children. These data also suggest bacterial markers for the early diagnosis of T1D. In addition, bacteria that negatively correlated with the autoimmune state may prove to be useful in the prevention of autoimmunity development in high-risk children.},
copyright = {2011 International Society for Microbial Ecology},
journal = {The ISME Journal},
language = {en},
number = {1}
}
@article{goffauFecalMicrobiotaComposition2013,
title = {Fecal {{Microbiota Composition Differs Between Children With}} {$\beta$}-{{Cell Autoimmunity}} and {{Those Without}}},
author = {de Goffau, Marcus C. and Luopaj{\"a}rvi, Kristiina and Knip, Mikael and Ilonen, Jorma and Ruohtula, Terhi and H{\"a}rk{\"o}nen, Taina and Orivuori, Laura and Hakala, Saara and Welling, Gjalt W. and Harmsen, Hermie J. and Vaarala, Outi},
year = {2013},
month = apr,
volume = {62},
pages = {1238--1244},
publisher = {{American Diabetes Association}},
issn = {0012-1797, 1939-327X},
doi = {10.2337/db12-0526},
abstract = {The role of the intestinal microbiota as a regulator of autoimmune diabetes in animal models is well-established, but data on human type 1 diabetes are tentative and based on studies including only a few study subjects. To exclude secondary effects of diabetes and HLA risk genotype on gut microbiota, we compared the intestinal microbiota composition in children with at least two diabetes-associated autoantibodies (n = 18) with autoantibody-negative children matched for age, sex, early feeding history, and HLA risk genotype using pyrosequencing. Principal component analysis indicated that a low abundance of lactate-producing and butyrate-producing species was associated with {$\beta$}-cell autoimmunity. In addition, a dearth of the two most dominant Bifidobacterium species, Bifidobacterium adolescentis and Bifidobacterium pseudocatenulatum, and an increased abundance of the Bacteroides genus were observed in the children with {$\beta$}-cell autoimmunity. We did not find increased fecal calprotectin or IgA as marker of inflammation in children with {$\beta$}-cell autoimmunity. Functional studies related to the observed alterations in the gut microbiome are warranted because the low abundance of bifidobacteria and butyrate-producing species could adversely affect the intestinal epithelial barrier function and inflammation, whereas the apparent importance of the Bacteroides genus in development of type 1 diabetes is insufficiently understood.},
chapter = {Original Research},
copyright = {\textcopyright{} 2013 by the American Diabetes Association.. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.},
journal = {Diabetes},
language = {en},
number = {4},
pmid = {23274889}
}
@article{goodmanExtensivePersonalHuman2011,
title = {Extensive Personal Human Gut Microbiota Culture Collections Characterized and Manipulated in Gnotobiotic Mice},
author = {Goodman, Andrew L. and Kallstrom, George and Faith, Jeremiah J. and Reyes, Alejandro and Moore, Aimee and Dantas, Gautam and Gordon, Jeffrey I.},
year = {2011},
month = apr,
volume = {108},