Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jul 15, 2021; 12(7): 1070-1080
Published online Jul 15, 2021. doi: 10.4239/wjd.v12.i7.1070
Multi-omics: Opportunities for research on mechanism of type 2 diabetes mellitus
Shuai Wang, Hui Yong, Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
Xiao-Dong He, Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
ORCID number: Shuai Wang (0000-0003-3533-8565); Hui Yong (0000-0001-7377-0266); Xiao-Dong He (0000-0002-5903-6003).
Author contributions: Wang S was responsible for drafting the article; Yong H and He XD made contributions to data acquisition; He XD contributed to conception and provided final approval of the version of the article to be published.
Supported by Grant from International Joint Usage/Research Center, the Institute of Medical Science, the University of Tokyo, No. New-2020-K2012; and Open Project of Shandong Provincial Key Laboratory of Infection and Immunity, No. 2.
Conflict-of-interest statement: The authors declare no conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xiao-Dong He, PhD, Lecturer, Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan 250012, Shandong Province, China. xiaodong.he@sdu.edu.cn
Received: January 24, 2021
Peer-review started: January 24, 2021
First decision: March 16, 2021
Revised: March 22, 2021
Accepted: May 22, 2021
Article in press: May 22, 2021
Published online: July 15, 2021

Abstract

Type 2 diabetes mellitus (T2DM) is a burdensome global disease. In-depth understanding of its mechanism will help to optimize diagnosis and treatment, which reduces the burden. Multi-omics research has unparalleled advantages in contributing to the overall understanding of the mechanism of this chronic metabolic disease. In the past two decades, the study of multi-omics on T2DM-related intestinal flora perturbation and plasma dyslipidemia has shown tremendous potential and is expected to achieve major breakthroughs. The regulation of intestinal flora in diabetic patients has been confirmed by multiple studies. The use of metagenomics, 16S RNA sequencing, and metabolomics has comprehensively identified the overall changes in the intestinal flora and the metabolic disturbances that could directly or indirectly participate in the intestinal flora-host interactions. Lipidomics combined with other “omics” has characterized lipid metabolism disorders in T2DM. The combined application and cross-validation of multi-omics can screen for dysregulation in T2DM, which will provide immense opportunities to understand the mechanisms behind T2DM.

Key Words: Type 2 diabetes mellitus, Gastrointestinal microbiome, Intestinal flora, Lipid metabolism disorders, Dyslipidemias, Metabolomics

Core Tip: The prospects of multi-omics in the study of the mechanisms of type 2 diabetes mellitus (T2DM)-related intestinal flora perturbation and plasma dyslipidemia are tremendous. The use of multi-omics has identified variations in T2DM intestinal flora composition and human-microbiota interactions. However, further sequencing is required, and the clinical application needs to be clarified and simplified. Multi-omics is also identifying T2DM lipid profiles, which will provide immense opportunities to understand the mechanisms of T2DM-related dyslipidemia.



INTRODUCTION

According to the World Health Organization, about 422 million people worldwide have diabetes and 1.6 million deaths are directly attributed to diabetes each year. Diabetes is a chronic, metabolic disease characterized by elevated blood glucose (or blood sugar) levels, which increases morbidity and mortality. When the body does not produce enough insulin or does not use it efficiently, diabetes manifests. The number of patients with diabetes is increasing, which expands the magnitude of the disease burden[1]. The most common type of diabetes is type 2 diabetes mellitus (T2DM)[2]. When genetics, age, and family history are fixed, reducing exposure to other known risk factors for T2DM using a variety of interventions and improving access to and quality of care decrease the incidence of T2DM and benefit patients with T2DM[3]. The effectiveness of these interventions depends largely on the understanding of the mechanism of T2DM. Due to the complexity of the mechanisms and causes of T2DM, traditional bench science has limitations. Multi-omics, including genomics, transcriptomics, proteomics, glycomics, metabolomics, epigenomics, ncRNomics, lipidomics, and interactomics, offers a fresh and exciting conceptual lens that will aid scientists to comprehensively and systematically understand the physiological processes and regulatory mechanisms of T2DM[4].

ADVANTAGES OF MULTI-OMICS ON STUDY OF MECHANISM OF T2DM

Since the concept of genomes and genomics was introduced, “omics” research has profoundly affected systems biology discoveries. Multi-omics research enables researchers to identify the differences in genes, proteins, and metabolites that lead to understanding overall functional disturbances in diseases, including T2DM.

Using multi-omics, our previous research[5,6] described the intestinal flora alterations and the plasma protein metabolic profile perturbations in the Zucker diabetic fatty rat, which is a spontaneous T2DM animal model commonly used to develop drugs for treating diabetes[7-10]. The altered intestinal microbiota and differentially expressed proteins and metabolites clearly distinguished the treatment group [T: Zucker leptin receptor gene-deficient rats (fa/fa) treated by Purina #5008] from the control group [C: basic diet-fed litter mate wild-type controls (fa/+)]. This provided an important reference for screening and verifying T2DM by utilizing intestinal flora and plasma biomarkers. Using “omics” techniques, the increased levels of glycated hemoglobin, ceruloplasmin, triacylglycerols, diacylglycerols, phosphatidylethanolamines, etc. in plasma/urine have been verified in the human population. They are gradually being introduced in the early diagnosis of diabetes and the prediction of serious adverse complications[11-13].

Further functional analysis of the differential molecules using multi-omics revealed the pathophysiological mechanism of T2DM (Figure 1). The intestinal flora and the host exhibit similar features that focus on oxidative stress, insulin resistance, and metabolic disorders. This data confirmed previous T2DM mechanism research[14-16] and provided insight into the overall levels of molecules.

Figure 1
Figure 1 Multi-omics verification of the mechanism of type 2 diabetes mellitus. A: Schematic diagram of multi-omics verification; B: Conceptual diagram of the Zucker diabetic fatty rat modeling process; C: Body weight (left) and blood sugar (right) before and after basic diet feeding in B; D: Fecal 16S rRNA sequencing biomarkers; E: Genomic functions of the disturbed intestinal flora; F: Overview of the complete metabolism in the two biological system; G: Protein-protein interaction network of plasma differentially expressed proteins (DEPs); H: Functional enrichment of DEPs; I: Visualization of G and H. The cross-validation of intestinal flora and the plasma proteome further emphasizes that oxidative stress, insulin resistance, energy intake and consumption imbalances, and lipid metabolism disorders play an important part in the occurrence of type 2 diabetes mellitus (T2DM). Dyslipidemia may be a major hub for the in vivo and in vitro changes in T2DM (unpublished data). The data used in the figure are our published and public data in open access journals, which are displayed after further analysis. C and G: Citation: Wang S, Lu Z, Wang Y, Zhang T, He X. Metalloproteins and apolipoprotein C: candidate plasma biomarkers of T2DM screened by comparative proteomics and lipidomics in ZDF rats. Nutr Metab (Lond) 2020; 17: 66. Copyright ©The Author(s) 2020. Published by Springer Nature[6]. T: Zucker leptin receptor gene-deficient rats (fa/fa) treated by Purina #5008 for 3 wk; C: Basic diet-fed litter mate wild-type controls (fa/+); DEPs: Differentially expressed proteins; T2DM: Type 2 diabetes mellitus.
MULTI-OMICS AND T2DM-RELATED INTESTINAL FLORA DISTURBANCE

Multi-omics aided the dramatic discovery that diabetes was associated with the intestinal flora. The altered microbiota observed in genetically obese mice[17] or people[18-20] is sufficient to promote increased adiposity in lean mice that receive a microbiota transplant. Germ-free mice, which lack a microbiota, have reduced adiposity and improved tolerance to glucose and insulin when compared to their counterparts[21]. They are also free from diet-induced obesity when fed a Western-style diet[22-24]. Taken together, the correlation between intestinal flora, obesity, and diabetes demonstrates that the microbiota contributes to the regulation of adiposity and T2DM[25]. However, the precise mechanism is still not clear.

Increased metagenomics data (usually 16S RNA sequencing) suggest that the extent of biodiversity within an ecosystem can be an important mechanism and serve as a measure of stability and robustness. In other words, a reduction in gut microbiome diversity and richness is linked to susceptibility to T2DM[26,27]. The complex composition of the intestinal flora can quickly change in response to a diverse diet, while simpler flora composition can only interact with specific diets and increase the vulnerability of the intestinal tract[28]. In T2DM rats and patients, the proportion of Firmicutes decreased and that of Bacteroidetes increased[29]. This ratio can be used as a simple indicator of intestinal flora diversity in T2DM. Although metagenomics has an irreplaceable advantage in T2DM-related intestinal flora studies, sequencing depth and defects in methods still need to be paid more attention. 16S RNA sequencing enables taxonomic identification to at least the family level but is rarely able to make a distinction between different strains of the same species or related species[30]. Salmonella, Escherichia coli, and Shigella would be identified by the same sequence, yet their significance in T2DM may vary.

Although the intestinal flora ratios may be important, the metabolic function of the intestinal flora on nutrients and food likely plays a more significant role[31,32]. Metabolites of the intestinal flora are involved in numerous functions. They can affect the absorption of nutrients (bile acid metabolism is closely related to lipid absorption[33,34]), and they can be absorbed to provide nutrients for the host (carbohydrates are fermented to form short-chain fatty acids to provide energy for the host[35-37]). Metabolites can be signaling molecules to regulate inflammation and immunity inter- or intra-intestinally. Bacterial L-tryptophan metabolites enhance the secretion of glucagon-like peptide-1[38-40]. Endotoxins (lipopolysaccharides) induce inflammation[41] and limit the autoimmune response[42,43]. Many metabolites produced due to the interaction of numerous species, the allocation of resources, and the dynamic response to perturbation within the gut may serve as potential T2DM biomarkers[44]. Table 1 briefly summarizes related gut microbiota metabolites and their interactions with the host in diabetes. The complementation of intestinal metabolomics and metagenomics will enhance the research on T2DM-related intestinal flora disturbance[45].

Table 1 Intestinal flora-related metabolites and their host interaction mechanism.
Metabolites
Potential interaction mechanism between intestinal flora-related metabolites and the host
Bile acidsPromotes fat absorption; serves as signaling molecule [acts with G-protein-coupled bile acid receptor 1 (Gpbar1, TGR5) and the bile acid receptor FXR]; limits the autoimmune response
SCFA
Acetate and butyrateActs as histone deacetylase inhibitors; ameliorates inflammation
PropionateParticipates in carbohydrate esterification
ValericProvides calories; affects inflammation; enteroendocrine regulation through G-protein-coupled receptors (e.g., GPR41, GPR43)
IndolePromotes the function of the intestinal cell epithelial barrier; enhances the secretion of glucagon-like peptide-1 (GLP-1)
Endotoxins (LPS)Induces inflammation; limits the autoimmune response
H2SDestroys the intestinal barrier function
TMAInterferes with metabolism

Discovering the interactions of the intestinal microbiota under complex conditions, such as diet[46], drugs[47], genetic background of the host[48] and colonizing flora[49], modification of the intestinal flora through antibiotics[50-54], probiotics, prebiotics[55-57], and fecal microbiota transplantation, and causal relationships in people and precision treatment[58] will become more accessible using multi-omics. The potential of understanding the mechanisms of diabetes using multi-omics is tremendous. However, the systematic examination of T2DM-related colonic flora studies in therapeutic and clinical application must still be completed.

An interesting example of fully understanding the mechanism of intestinal bacteria is Akkermansia muciniphila (A. muciniphila). A. muciniphila is a mucosal-dwelling anaerobe and the only known member of its genus. Animal and human data have indicated an inverse correlation between the intestinal abundance of A. muciniphila and obesity, dyslipidemia, and T2DM[59-61]. A. muciniphila supplementation restores epithelial mucosal integrity, reduces weight gain and fat accumulation, improves glucose tolerance, and reduces inflammation and metabolic endotoxemia in animal models. It may be a potential treatment for T2DM. However, A. muciniphila levels are increased in patients with Parkinson’s disease, multiple sclerosis, or Alzheimer’s disease[62-64], suggesting that this bacterium may have unforeseen harmful effects on the nervous system. Pure culture, sterilization, and component extraction can reduce this risk[65], which is consistent with the recently proposed concept of culturomics[66]. In short, through multi-omics it is likely that major breakthroughs in the study of the mechanism of T2DM-related intestinal flora perturbation will be made and that these breakthroughs will be gradually applied in the clinic.

MULTI-OMICS AND T2DM-RELATED DYSLIPIDEMIA

While intestinal flora disorder is a diet-related in vitro regulatory mechanism of T2DM, glucose and lipid metabolism disorders and insulin resistance are in vivo mechanisms[67]. In addition to glycomics[68,69], we want to emphasize the role of lipidomics in T2DM-related dyslipidemia. Dyslipidemia in T2DM, characterized by a high concentration of triglycerides, low concentration of high-density lipoprotein cholesterol, and increased concentration of small, dense low-density lipoprotein cholesterol particles, is associated with insulin resistance. It is also one of the major risk factors for cardiovascular disease in patients with diabetes[70]. Although active control of triglycerides and low-density lipoprotein cholesterol can delay the progression of T2DM and reduce the risk of adverse cardiovascular outcomes in patients, interventions to increase high-density lipoprotein cholesterol have had little success. Niacin was thought to raise high-density lipoprotein cholesterol and was used to control diabetic dyslipidemia previously. However, it was proven to be ineffective and removed from the treatment guidelines[71-75]. Lipidomics, the systematic analysis of lipid composition and expression changes, can intensify the understanding of lipid metabolism alterations in T2DM. Recent population lipidomics data revealed that the T2DM-related lipid profile included decreased lysophospholipids, phosphatidylcholines, sphingomyelins, and cholesterol esters and increased triacylglycerols, diacylglycerols, and phosphatidylethanolamines[76-78]. Although these indicators can be used as potential biomarkers to predict the risk of T2DM, its prediction for a particular disease has not yet been verified. Balgoma et al[79] compared the lipid profiles from patients with non-alcoholic fatty liver disease, cardiovascular incidents, hepatocellular carcinoma, and T2DM. They noted that the upregulation of triacylglycerols, palmitic acid, palmitoleic acid, stearic acid and oleic acid is a fingerprint of liver X receptor-mediated lipogenesis in the liver. To thoroughly understand the mechanisms of T2DM-related lipid metabolism disorders, the utilization of lipidomics, and the integration of databases will undoubtedly achieve this goal.

CONCLUSION

Multi-omic studies have provided new breakthroughs and directions to guide traditional molecular biology research. The expansion of “omics” data and the continuous advancement of bioinformatics analysis technology will surely continue the advancement of our knowledge on the mechanisms of T2DM, especially on intestinal flora perturbation and dyslipidemia.

ACKNOWLEDGEMENTS

The authors want to thank Ms. Wang SE and Ms. Liu S for their suggestions and discussions during the writing process.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Endocrinology and metabolism

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Mazlan M S-Editor: Gao CC L-Editor: Wang TQ P-Editor: Ma YJ

References
1.  Saeedi P, Salpea P, Karuranga S, Petersohn I, Malanda B, Gregg EW, Unwin N, Wild SH, Williams R. Mortality attributable to diabetes in 20-79 years old adults, 2019 estimates: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract. 2020;162:108086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 281]  [Article Influence: 70.3]  [Reference Citation Analysis (1)]
2.  Cole AR, Astell A, Green C, Sutherland C. Molecular connexions between dementia and diabetes. Neurosci Biobehav Rev. 2007;31:1046-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 103]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
3.  Chatterjee S, Khunti K, Davies MJ. Type 2 diabetes. Lancet. 2017;389:2239-2251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1234]  [Cited by in F6Publishing: 1403]  [Article Influence: 200.4]  [Reference Citation Analysis (1)]
4.  Wang N, Zhu F, Chen L, Chen K. Proteomics, metabolomics and metagenomics for type 2 diabetes and its complications. Life Sci. 2018;212:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (1)]
5.  Wang Y, Ouyang M, Gao X, Wang S, Fu C, Zeng J, He X. Phocea, Pseudoflavonifractor and Lactobacillus intestinalis: Three Potential Biomarkers of Gut Microbiota That Affect Progression and Complications of Obesity-Induced Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes. 2020;13:835-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
6.  Wang S, Lu Z, Wang Y, Zhang T, He X. Metalloproteins and apolipoprotein C: candidate plasma biomarkers of T2DM screened by comparative proteomics and lipidomics in ZDF rats. Nutr Metab (Lond). 2020;17:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
7.  Ohneda M, Inman LR, Unger RH. Caloric restriction in obese pre-diabetic rats prevents beta-cell depletion, loss of beta-cell GLUT 2 and glucose incompetence. Diabetologia. 1995;38:173-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 81]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
8.  Cefalu WT. Animal models of type 2 diabetes: clinical presentation and pathophysiological relevance to the human condition. ILAR J. 2006;47:186-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 125]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
9.  Nugent DA, Smith DM, Jones HB. A review of islet of Langerhans degeneration in rodent models of type 2 diabetes. Toxicol Pathol. 2008;36:529-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
10.  Al-Awar A, Kupai K, Veszelka M, Szűcs G, Attieh Z, Murlasits Z, Török S, Pósa A, Varga C. Experimental Diabetes Mellitus in Different Animal Models. J Diabetes Res. 2016;2016:9051426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 149]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
11.  Hameed A, Mojsak P, Buczynska A, Suleria HAR, Kretowski A, Ciborowski M. Altered Metabolome of Lipids and Amino Acids Species: A Source of Early Signature Biomarkers of T2DM. J Clin Med. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
12.  Ge S, Wang Y, Song M, Li X, Yu X, Wang H, Wang J, Zeng Q, Wang W. Type 2 Diabetes Mellitus: Integrative Analysis of Multiomics Data for Biomarker Discovery. OMICS. 2018;22:514-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
13.  Kim SW, Choi JW, Yun JW, Chung IS, Cho HC, Song SE, Im SS, Song DK. Proteomics approach to identify serum biomarkers associated with the progression of diabetes in Korean patients with abdominal obesity. PLoS One. 2019;14:e0222032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
14.  Wada J, Makino H. Inflammation and the pathogenesis of diabetic nephropathy. Clin Sci (Lond). 2013;124:139-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 447]  [Cited by in F6Publishing: 519]  [Article Influence: 47.2]  [Reference Citation Analysis (0)]
15.  Mohamed J, Nazratun Nafizah AH, Zariyantey AH, Budin SB. Mechanisms of Diabetes-Induced Liver Damage: The role of oxidative stress and inflammation. Sultan Qaboos Univ Med J. 2016;16:e132-e141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 227]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
16.  Rehman K, Akash MSH. Mechanism of Generation of Oxidative Stress and Pathophysiology of Type 2 Diabetes Mellitus: How Are They Interlinked? J Cell Biochem. 2017;118:3577-3585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 287]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
17.  Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7820]  [Cited by in F6Publishing: 8111]  [Article Influence: 477.1]  [Reference Citation Analysis (1)]
18.  Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto JM, Kennedy S, Leonard P, Li J, Burgdorf K, Grarup N, Jørgensen T, Brandslund I, Nielsen HB, Juncker AS, Bertalan M, Levenez F, Pons N, Rasmussen S, Sunagawa S, Tap J, Tims S, Zoetendal EG, Brunak S, Clément K, Doré J, Kleerebezem M, Kristiansen K, Renault P, Sicheritz-Ponten T, de Vos WM, Zucker JD, Raes J, Hansen T;  MetaHIT consortium; Bork P, Wang J, Ehrlich SD, Pedersen O. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2727]  [Cited by in F6Publishing: 2791]  [Article Influence: 253.7]  [Reference Citation Analysis (1)]
19.  Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R, Newgard CB, Heath AC, Gordon JI. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2415]  [Cited by in F6Publishing: 2480]  [Article Influence: 225.5]  [Reference Citation Analysis (0)]
20.  Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, Spector TD, Clark AG, Ley RE. Human genetics shape the gut microbiome. Cell. 2014;159:789-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1911]  [Cited by in F6Publishing: 1942]  [Article Influence: 215.8]  [Reference Citation Analysis (0)]
21.  Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 2004;101:15718-15723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4530]  [Cited by in F6Publishing: 4117]  [Article Influence: 205.9]  [Reference Citation Analysis (4)]
22.  Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA. 2007;104:979-984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1778]  [Cited by in F6Publishing: 1764]  [Article Influence: 103.8]  [Reference Citation Analysis (0)]
23.  Rabot S, Membrez M, Bruneau A, Gérard P, Harach T, Moser M, Raymond F, Mansourian R, Chou CJ. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 2010;24:4948-4959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 382]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
24.  Ding S, Chi MM, Scull BP, Rigby R, Schwerbrock NM, Magness S, Jobin C, Lund PK. High-fat diet: bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS One. 2010;5:e12191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 476]  [Cited by in F6Publishing: 496]  [Article Influence: 35.4]  [Reference Citation Analysis (0)]
25.  Navab-Moghadam F, Sedighi M, Khamseh ME, Alaei-Shahmiri F, Talebi M, Razavi S, Amirmozafari N. The association of type II diabetes with gut microbiota composition. Microb Pathog. 2017;110:630-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 79]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
26.  Cardinale BJ, Duffy JE, Gonzalez A, Hooper DU, Perrings C, Venail P, Narwani A, Mace GM, Tilman D, Wardle DA, Kinzig AP, Daily GC, Loreau M, Grace JB, Larigauderie A, Srivastava DS, Naeem S. Biodiversity loss and its impact on humanity. Nature. 2012;486:59-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3783]  [Cited by in F6Publishing: 2345]  [Article Influence: 195.4]  [Reference Citation Analysis (0)]
27.  Vallianou N, Stratigou T, Christodoulatos GS, Dalamaga M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr Obes Rep. 2019;8:317-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 141]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
28.  Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis JD. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4098]  [Cited by in F6Publishing: 4123]  [Article Influence: 317.2]  [Reference Citation Analysis (0)]
29.  Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, Al-Soud WA, Sørensen SJ, Hansen LH, Jakobsen M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1783]  [Cited by in F6Publishing: 1889]  [Article Influence: 134.9]  [Reference Citation Analysis (0)]
30.  Cirstea M, Radisavljevic N, Finlay BB. Good Bug, Bad Bug: Breaking through Microbial Stereotypes. Cell Host Microbe. 2018;23:10-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
31.  Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, Tuohy K. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57:1-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 993]  [Cited by in F6Publishing: 1265]  [Article Influence: 180.7]  [Reference Citation Analysis (0)]
32.  Pascale A, Marchesi N, Marelli C, Coppola A, Luzi L, Govoni S, Giustina A, Gazzaruso C. Microbiota and metabolic diseases. Endocrine. 2018;61:357-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 233]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
33.  Kawamata Y, Fujii R, Hosoya M, Harada M, Yoshida H, Miwa M, Fukusumi S, Habata Y, Itoh T, Shintani Y, Hinuma S, Fujisawa Y, Fujino M. A G protein-coupled receptor responsive to bile acids. J Biol Chem. 2003;278:9435-9440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1060]  [Cited by in F6Publishing: 1112]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
34.  Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov. 2008;7:678-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 869]  [Cited by in F6Publishing: 921]  [Article Influence: 57.6]  [Reference Citation Analysis (0)]
35.  McNeil NI. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr. 1984;39:338-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 420]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
36.  Bergman EN. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev. 1990;70:567-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1559]  [Cited by in F6Publishing: 1541]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
37.  Mandaliya DK, Seshadri S. Short Chain Fatty Acids, pancreatic dysfunction and type 2 diabetes. Pancreatology. 2019;19:280-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
38.  Miele L, Giorgio V, Alberelli MA, De Candia E, Gasbarrini A, Grieco A. Impact of Gut Microbiota on Obesity, Diabetes, and Cardiovascular Disease Risk. Curr Cardiol Rep. 2015;17:120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 96]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
39.  de Mello VD, Paananen J, Lindström J, Lankinen MA, Shi L, Kuusisto J, Pihlajamäki J, Auriola S, Lehtonen M, Rolandsson O, Bergdahl IA, Nordin E, Ilanne-Parikka P, Keinänen-Kiukaanniemi S, Landberg R, Eriksson JG, Tuomilehto J, Hanhineva K, Uusitupa M. Indolepropionic acid and novel lipid metabolites are associated with a lower risk of type 2 diabetes in the Finnish Diabetes Prevention Study. Sci Rep. 2017;7:46337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 200]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
40.  Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T, Yin Y. Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism. Front Cell Infect Microbiol. 2018;8:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 752]  [Cited by in F6Publishing: 672]  [Article Influence: 112.0]  [Reference Citation Analysis (0)]
41.  Caesar R, Reigstad CS, Bäckhed HK, Reinhardt C, Ketonen M, Lundén GÖ, Cani PD, Bäckhed F. Gut-derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut. 2012;61:1701-1707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 211]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
42.  Saito T, Hayashida H, Furugen R. Comment on: Cani et al (2007) Metabolic endotoxemia initiates obesity and insulin resistance: Diabetes 56:1761-1772. Diabetes. 2007;56:e20; author reply e21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
43.  Erridge C, Attina T, Spickett CM, Webb DJ. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflammation. Am J Clin Nutr. 2007;86:1286-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 518]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
44.  Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1187]  [Cited by in F6Publishing: 1288]  [Article Influence: 161.0]  [Reference Citation Analysis (0)]
45.  Peng W, Huang J, Yang J, Zhang Z, Yu R, Fayyaz S, Zhang S, Qin YH. Integrated 16S rRNA Sequencing, Metagenomics, and Metabolomics to Characterize Gut Microbial Composition, Function, and Fecal Metabolic Phenotype in Non-obese Type 2 Diabetic Goto-Kakizaki Rats. Front Microbiol. 2019;10:3141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
46.  David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5625]  [Cited by in F6Publishing: 5986]  [Article Influence: 544.2]  [Reference Citation Analysis (0)]
47.  Rodriguez J, Hiel S, Delzenne NM. Metformin: old friend, new ways of action-implication of the gut microbiome? Curr Opin Clin Nutr Metab Care. 2018;21:294-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
48.  Pryor R, Norvaisas P, Marinos G, Best L, Thingholm LB, Quintaneiro LM, De Haes W, Esser D, Waschina S, Lujan C, Smith RL, Scott TA, Martinez-Martinez D, Woodward O, Bryson K, Laudes M, Lieb W, Houtkooper RH, Franke A, Temmerman L, Bjedov I, Cochemé HM, Kaleta C, Cabreiro F. Host-Microbe-Drug-Nutrient Screen Identifies Bacterial Effectors of Metformin Therapy. Cell 2019; 178: 1299-1312. e29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 154]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
49.  Lee SM, Donaldson GP, Mikulski Z, Boyajian S, Ley K, Mazmanian SK. Bacterial colonization factors control specificity and stability of the gut microbiota. Nature. 2013;501:426-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 381]  [Cited by in F6Publishing: 413]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
50.  Membrez M, Blancher F, Jaquet M, Bibiloni R, Cani PD, Burcelin RG, Corthesy I, Macé K, Chou CJ. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J. 2008;22:2416-2426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 368]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
51.  Chou CJ, Membrez M, Blancher F. Gut decontamination with norfloxacin and ampicillin enhances insulin sensitivity in mice. Nestle Nutr Workshop Ser Pediatr Program. 2008;62:127-137; discussion 137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
52.  Han J, Lin H, Huang W. Modulating gut microbiota as an anti-diabetic mechanism of berberine. Med Sci Monit. 2011;17:RA164-RA167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
53.  Thuny F, Richet H, Casalta JP, Angelakis E, Habib G, Raoult D. Vancomycin treatment of infective endocarditis is linked with recently acquired obesity. PLoS One. 2010;5:e9074.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 116]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
54.  Hernández E, Bargiela R, Diez MS, Friedrichs A, Pérez-Cobas AE, Gosalbes MJ, Knecht H, Martínez-Martínez M, Seifert J, von Bergen M, Artacho A, Ruiz A, Campoy C, Latorre A, Ott SJ, Moya A, Suárez A, Martins dos Santos VA, Ferrer M. Functional consequences of microbial shifts in the human gastrointestinal tract linked to antibiotic treatment and obesity. Gut Microbes. 2013;4:306-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
55.  Yadav H, Jain S, Sinha PR. Antidiabetic effect of probiotic dahi containing Lactobacillus acidophilus and Lactobacillus casei in high fructose fed rats. Nutrition. 2007;23:62-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 337]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
56.  Moroti C, Souza Magri LF, de Rezende Costa M, Cavallini DC, Sivieri K. Effect of the consumption of a new symbiotic shake on glycemia and cholesterol levels in elderly people with type 2 diabetes mellitus. Lipids Health Dis. 2012;11:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 149]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
57.  Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012;28:539-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 385]  [Article Influence: 29.6]  [Reference Citation Analysis (0)]
58.  Leustean AM, Ciocoiu M, Sava A, Costea CF, Floria M, Tarniceriu CC, Tanase DM. Implications of the Intestinal Microbiota in Diagnosing the Progression of Diabetes and the Presence of Cardiovascular Complications. J Diabetes Res. 2018;2018:5205126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
59.  Derrien M, Belzer C, de Vos WM. Akkermansia muciniphila and its role in regulating host functions. Microb Pathog. 2017;106:171-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 604]  [Article Influence: 75.5]  [Reference Citation Analysis (0)]
60.  Zhai Q, Feng S, Arjan N, Chen W. A next generation probiotic, Akkermansia muciniphila. Crit Rev Food Sci Nutr. 2019;59:3227-3236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 192]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
61.  Xu Y, Wang N, Tan HY, Li S, Zhang C, Feng Y. Function of Akkermansia muciniphila in Obesity: Interactions With Lipid Metabolism, Immune Response and Gut Systems. Front Microbiol. 2020;11:219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 223]  [Article Influence: 55.8]  [Reference Citation Analysis (0)]
62.  Heintz-Buschart A, Pandey U, Wicke T, Sixel-Döring F, Janzen A, Sittig-Wiegand E, Trenkwalder C, Oertel WH, Mollenhauer B, Wilmes P. The nasal and gut microbiome in Parkinson's disease and idiopathic rapid eye movement sleep behavior disorder. Mov Disord. 2018;33:88-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 330]  [Article Influence: 47.1]  [Reference Citation Analysis (0)]
63.  Hill-Burns EM, Debelius JW, Morton JT, Wissemann WT, Lewis MR, Wallen ZD, Peddada SD, Factor SA, Molho E, Zabetian CP, Knight R, Payami H. Parkinson's disease and Parkinson's disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32:739-749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 459]  [Cited by in F6Publishing: 543]  [Article Influence: 77.6]  [Reference Citation Analysis (0)]
64.  Cekanaviciute E, Yoo BB, Runia TF, Debelius JW, Singh S, Nelson CA, Kanner R, Bencosme Y, Lee YK, Hauser SL, Crabtree-Hartman E, Sand IK, Gacias M, Zhu Y, Casaccia P, Cree BAC, Knight R, Mazmanian SK, Baranzini SE. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc Natl Acad Sci USA. 2017;114:10713-10718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 502]  [Cited by in F6Publishing: 593]  [Article Influence: 84.7]  [Reference Citation Analysis (0)]
65.  Plovier H, Everard A, Druart C, Depommier C, Van Hul M, Geurts L, Chilloux J, Ottman N, Duparc T, Lichtenstein L, Myridakis A, Delzenne NM, Klievink J, Bhattacharjee A, van der Ark KC, Aalvink S, Martinez LO, Dumas ME, Maiter D, Loumaye A, Hermans MP, Thissen JP, Belzer C, de Vos WM, Cani PD. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med. 2017;23:107-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 967]  [Cited by in F6Publishing: 1180]  [Article Influence: 147.5]  [Reference Citation Analysis (0)]
66.  Kambouris ME, Pavlidis C, Skoufas E, Arabatzis M, Kantzanou M, Velegraki A, Patrinos GP. Culturomics: A New Kid on the Block of OMICS to Enable Personalized Medicine. OMICS. 2018;22:108-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
67.  Tomkin GH. Dyslipidaemia--hepatic and intestinal cross-talk. Atheroscler Suppl. 2010;11:5-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
68.  Liu J, Dolikun M, Štambuk J, Trbojević-Akmačić I, Zhang J, Wang H, Meng X, Razdorov G, Menon D, Zheng D, Wu L, Wang Y, Song M, Lauc G, Wang W. Glycomics for Type 2 Diabetes Biomarker Discovery: Promise of Immunoglobulin G Subclass-Specific Fragment Crystallizable N-glycosylation in the Uyghur Population. OMICS. 2019;23:640-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
69.  Kunej T. Rise of Systems Glycobiology and Personalized Glycomedicine: Why and How to Integrate Glycomics with Multiomics Science? OMICS. 2019;23:615-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
70.  Mooradian AD. Dyslipidemia in type 2 diabetes mellitus. Nat Clin Pract Endocrinol Metab. 2009;5:150-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in F6Publishing: 496]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
71.  HPS2-THRIVE Collaborative Group; Landray MJ, Haynes R, Hopewell JC, Parish S, Aung T, Tomson J, Wallendszus K, Craig M, Jiang L, Collins R, Armitage J. Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med. 2014;371:203-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1131]  [Cited by in F6Publishing: 1108]  [Article Influence: 110.8]  [Reference Citation Analysis (0)]
72.  Tuteja S, Rader DJ. Dyslipidaemia: cardiovascular prevention--end of the road for niacin? Nat Rev Endocrinol. 2014;10:646-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
73.  Mani P, Rohatgi A. Niacin Therapy, HDL Cholesterol, and Cardiovascular Disease: Is the HDL Hypothesis Defunct? Curr Atheroscler Rep. 2015;17:43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
74.  Kent S, Haynes R, Hopewell JC, Parish S, Gray A, Landray MJ, Collins R, Armitage J, Mihaylova B;  HPS2-THRIVE Collaborative Group. Effects of Vascular and Nonvascular Adverse Events and of Extended-Release Niacin With Laropiprant on Health and Healthcare Costs. Circ Cardiovasc Qual Outcomes. 2016;9:348-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
75.  Haynes R, Valdes-Marquez E, Hopewell JC, Chen F, Li J, Parish S, Landray MJ, Armitage J;  HPS2-THRIVE Collaborative Group;  HPS2-THRIVE Writing Committee members;  HPS2-THRIVE Steering Committee members. Serious Adverse Effects of Extended-release Niacin/Laropiprant: Results From the Heart Protection Study 2-Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) Trial. Clin Ther. 2019;41:1767-1777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
76.  Razquin C, Toledo E, Clish CB, Ruiz-Canela M, Dennis C, Corella D, Papandreou C, Ros E, Estruch R, Guasch-Ferré M, Gómez-Gracia E, Fitó M, Yu E, Lapetra J, Wang D, Romaguera D, Liang L, Alonso-Gómez A, Deik A, Bullo M, Serra-Majem L, Salas-Salvadó J, Hu FB, Martínez-González MA. Plasma Lipidomic Profiling and Risk of Type 2 Diabetes in the PREDIMED Trial. Diabetes Care. 2018;41:2617-2624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 125]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
77.  Suvitaival T, Bondia-Pons I, Yetukuri L, Pöhö P, Nolan JJ, Hyötyläinen T, Kuusisto J, Orešič M. Lipidome as a predictive tool in progression to type 2 diabetes in Finnish men. Metabolism. 2018;78:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 95]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
78.  Mousa A, Naderpoor N, Mellett N, Wilson K, Plebanski M, Meikle PJ, de Courten B. Lipidomic profiling reveals early-stage metabolic dysfunction in overweight or obese humans. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864:335-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
79.  Balgoma D, Pettersson C, Hedeland M. Common Fatty Markers in Diseases with Dysregulated Lipogenesis. Trends Endocrinol Metab. 2019;30:283-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]