286 results on '"Andrew D. Patterson"'
Search Results
52. Data from Aberrant Lipid Metabolism in Hepatocellular Carcinoma Revealed by Plasma Metabolomics and Lipid Profiling
- Author
-
Jeffrey R. Idle, Jean-François Dufour, Frank J. Gonzalez, Thomas Pabst, Kristopher W. Krausz, Christian Lanz, Diren Beyoğlu, Olivier Maurhofer, and Andrew D. Patterson
- Abstract
There has been limited analysis of the effects of hepatocellular carcinoma (HCC) on liver metabolism and circulating endogenous metabolites. Here, we report the findings of a plasma metabolomic investigation of HCC patients by ultraperformance liquid chromatography-electrospray ionization-quadrupole time-of-flight mass spectrometry (UPLC-ESI-QTOFMS), random forests machine learning algorithm, and multivariate data analysis. Control subjects included healthy individuals as well as patients with liver cirrhosis or acute myeloid leukemia. We found that HCC was associated with increased plasma levels of glycodeoxycholate, deoxycholate 3-sulfate, and bilirubin. Accurate mass measurement also indicated upregulation of biliverdin and the fetal bile acids 7α-hydroxy-3-oxochol-4-en-24-oic acid and 3-oxochol-4,6-dien-24-oic acid in HCC patients. A quantitative lipid profiling of patient plasma was also conducted by ultraperformance liquid chromatography-electrospray ionization-triple quadrupole mass spectrometry (UPLC-ESI-TQMS). By this method, we found that HCC was also associated with reduced levels of lysophosphocholines and in 4 of 20 patients with increased levels of lysophosphatidic acid [LPA(16:0)], where it correlated with plasma α-fetoprotein levels. Interestingly, when fatty acids were quantitatively profiled by gas chromatography-mass spectrometry (GC-MS), we found that lignoceric acid (24:0) and nervonic acid (24:1) were virtually absent from HCC plasma. Overall, this investigation illustrates the power of the new discovery technologies represented in the UPLC-ESI-QTOFMS platform combined with the targeted, quantitative platforms of UPLC-ESI-TQMS and GC-MS for conducting metabolomic investigations that can engender new insights into cancer pathobiology. Cancer Res; 71(21); 6590–600. ©2011 AACR.
- Published
- 2023
53. Data from Mass Spectrometry–Based Metabolomics Identifies Longitudinal Urinary Metabolite Profiles Predictive of Radiation-Induced Cancer
- Author
-
Andrew D. Patterson, James B. Mitchell, Frank J. Gonzalez, Kristopher W. Krausz, Angela Thetford, Anastasia L. Sowers, Miriam R. Anver, Gadisetti V.R. Chandramouli, and John A. Cook
- Abstract
Nonlethal exposure to ionizing radiation (IR) is a public concern due to its known carcinogenic effects. Although latency periods for IR-induced neoplasms are relatively long, the ability to detect cancer as early as possible is highly advantageous for effective therapeutic intervention. Therefore, we hypothesized that metabolites in the urine from mice exposed to total body radiation (TBI) would predict for the presence of cancer before a palpable mass was detected. In this study, we exposed mice to 0 or 5.4 Gy TBI, collected urine samples periodically over 1 year, and assayed urine metabolites by using mass spectrometry. Longitudinal data analysis within the first year post-TBI revealed that cancers, including hematopoietic, solid, and benign neoplasms, could be distinguished by unique urinary signatures as early as 3 months post-TBI. Furthermore, a distinction among different types of malignancies could be clearly delineated as early as 3 months post-TBI for hematopoietic neoplasms, 6 months for solid neoplasms, and by 1 year for benign neoplasms. Moreover, the feature profile for radiation-exposed mice 6 months post-TBI was found to be similar to nonirradiated control mice at 18 months, suggesting that TBI accelerates aging. These results demonstrate that urine feature profiles following TBI can identify cancers in mice prior to macroscopic detection, with important implications for the early diagnosis and treatment. Cancer Res; 76(6); 1569–77. ©2016 AACR.
- Published
- 2023
54. Supplementary Materials and Methods, Figures 1 - 10, Tables 1 - 4 from Noninvasive Urinary Metabolomic Profiling Identifies Diagnostic and Prognostic Markers in Lung Cancer
- Author
-
Curtis C. Harris, Frank J. Gonzalez, Emmanuel Hatzakis, Dickran G. Kazandjian, Katsuhiro Anami, Philip B. Smith, Jeffrey R. Idle, Peter G. Shields, Elise D. Bowman, Kristopher W. Krausz, Soumen K. Manna, Majda Haznadar, Andrew D. Patterson, and Ewy A. Mathé
- Abstract
PDF file - 1526KB, Supplementary Table 1 shows random forest analysis results for predictions of lung cancer status in the training set. Supplementary Table 2 shows associations with survival in the training set when the top four predictive metabolites are combined in all cases. Supplementary Table 3 shows associations with survival in the training set, stratified by self-reported race. Supplementary Table 4 shows intraclass correlation coefficients in the quantitated subset. Supplementary Figure 1 depicts workflow of the classification analysis. Supplementary Figure 2 depicts quality control assessment in the training set. Supplementary Figure 3 shows predictions of smoking status in the training set determined by random forest analysis and abundances of tobacco-related metabolites. Supplementary Figure 4 shows overlap of metabolites predictive of lung cancer status in the training set based on random forest analysis, stratified by gender, race and smoking status. Supplementary Figure 5 shows fragmentation patterns of top four predictive metabolites determined by tandem mass spectrometry. Supplementary Figure 6 depicts identification of creatine riboside by NMR. Supplementary Figure 7 shows diurnal effects on top four predictive metabolites. Supplementary Figure 8 shows top four predictive metabolite abundances stratified by smoking status. Supplementary Figure 9 shows Kaplan-Meier survival estimates in the training set depicted for the top four predictive metabolites in stages I-II and their combination. Supplementary Figure 10 shows metabolite abundances stratified by chemotherapy/radiation status and surgery status.
- Published
- 2023
55. Supplementary Figure S2 from Mass Spectrometry–Based Metabolomics Identifies Longitudinal Urinary Metabolite Profiles Predictive of Radiation-Induced Cancer
- Author
-
Andrew D. Patterson, James B. Mitchell, Frank J. Gonzalez, Kristopher W. Krausz, Angela Thetford, Anastasia L. Sowers, Miriam R. Anver, Gadisetti V.R. Chandramouli, and John A. Cook
- Abstract
Comparison of metabolite profiles of 5.4 Gy (IR) to 0 Gy (Control) using 525 samples (Control = 141 from 17 mice, IR = 384 from 51 mice).
- Published
- 2023
56. Supplementary Methods and References from Mass Spectrometry–Based Metabolomics Identifies Longitudinal Urinary Metabolite Profiles Predictive of Radiation-Induced Cancer
- Author
-
Andrew D. Patterson, James B. Mitchell, Frank J. Gonzalez, Kristopher W. Krausz, Angela Thetford, Anastasia L. Sowers, Miriam R. Anver, Gadisetti V.R. Chandramouli, and John A. Cook
- Abstract
Description of additional methods and procedures used in the study. Also includes Supplementary References.
- Published
- 2023
57. Supplementary Figure 2 from Aberrant Lipid Metabolism in Hepatocellular Carcinoma Revealed by Plasma Metabolomics and Lipid Profiling
- Author
-
Jeffrey R. Idle, Jean-François Dufour, Frank J. Gonzalez, Thomas Pabst, Kristopher W. Krausz, Christian Lanz, Diren Beyoğlu, Olivier Maurhofer, and Andrew D. Patterson
- Abstract
PDF file - 1.3MB
- Published
- 2023
58. Supplementary Table 1 from Aberrant Lipid Metabolism in Hepatocellular Carcinoma Revealed by Plasma Metabolomics and Lipid Profiling
- Author
-
Jeffrey R. Idle, Jean-François Dufour, Frank J. Gonzalez, Thomas Pabst, Kristopher W. Krausz, Christian Lanz, Diren Beyoğlu, Olivier Maurhofer, and Andrew D. Patterson
- Abstract
PDF file - 63K
- Published
- 2023
59. Supplementary Tables S1-S6 from Mass Spectrometry–Based Metabolomics Identifies Longitudinal Urinary Metabolite Profiles Predictive of Radiation-Induced Cancer
- Author
-
Andrew D. Patterson, James B. Mitchell, Frank J. Gonzalez, Kristopher W. Krausz, Angela Thetford, Anastasia L. Sowers, Miriam R. Anver, Gadisetti V.R. Chandramouli, and John A. Cook
- Abstract
Distribution of neoplasm types between Control (0 Gy) and irradiated (5.4 Gy) mice (excluding outliers) ; Number of feature samples for various groups at different time intervals within 1 year ; Retention times and m/z ratios of quality controls and their standard errors ; Summary of Random Forest Classification ; Significant features (FDR
- Published
- 2023
60. Itaconate promotes the differentiation of murine stress erythroid progenitors by increasing Nrf2 activity
- Author
-
Baiye Ruan, Yuanting Chen, Sara Trimidal, Imhoi Koo, Jingwei Cai, John Mcguigan, Molly A. Hall, Andrew D. Patterson, K. Sandeep Prabhu, and Robert F. Paulson
- Abstract
Steady state erythropoiesis produces new erythrocytes at a constant rate to replace senescent erythrocytes removed in the spleen and liver. Inflammation caused by infection or tissue damage skews bone marrow hematopoiesis, increasing myelopoiesis at the expense of steady state erythropoiesis. To compensate for the loss of production, stress erythropoiesis is induced. Stress erythropoiesis is highly conserved between mouse and human but utilizes a different strategy than steady state erythropoiesis. Inflammatory signals promote the proliferation of immature stress erythroid progenitors (SEPS), which in response to erythropoietin and other signals transition to stress erythroid progenitors committed to differentiation. Here we show that TNFα dependent signaling increases ROS in SEPs during the proliferation stage, however, blocking ROS production impairs their later differentiation. In addition to TNFα, nitric oxide dependent signaling drives the proliferation of stress erythroid progenitors and production of nitric oxide must be decreased so that the progenitor cells can differentiate. As progenitor cells transition to differentiation, increased production of the anti-inflammatory metabolite itaconate activates Nfe2l2 or Nrf2, which inhibits Nos2 expression, leading to decreased nitric oxide production. Mutation of Irg1, the enzyme that catalyzes the production of itaconate, causes a delayed recovery from inflammatory anemia induced by heat killedBrucella abortus. Loss of itaconate-dependent activation of Nrf2 is rescued in vivo by IL-10, which leads to activation of Nrf2 and differentiation. These data show that the differentiation of stress erythroid progenitors relies on a switch to an anti-inflammatory metabolism and increased expression of pro-resolving cytokines.Key points1.The transition to differentiation of stress erythroid progenitors requires anti-inflammatory signals.2.The anti-inflammatory metabolite itaconate and IL-10 increase Nrf2 activity to promote the differentiation of stress erythroid progenitors.
- Published
- 2023
61. Nitric oxide regulates metabolism in murine stress erythroid progenitors to promote recovery during inflammatory anemia
- Author
-
Baiye Ruan, Yuanting Chen, Sara Trimidal, Imhoi Koo, Fenghua Qian, Jingwei Cai, John Mcguigan, Molly A. Hall, Andrew D. Patterson, K. Sandeep Prabhu, and Robert F. Paulson
- Subjects
Article - Abstract
Inflammation skews bone marrow hematopoiesis increasing the production of myeloid effector cells at the expense of steady-state erythropoiesis. A compensatory stress erythropoiesis response is induced to maintain homeostasis until inflammation is resolved. In contrast to steady-state erythroid progenitors, stress erythroid progenitors (SEPs) utilize signals induced by inflammatory stimuli. However, the mechanistic basis for this is not clear. Here we reveal a nitric oxide (NO)-dependent regulatory network underlying two stages of stress erythropoiesis, namely proliferation, and the transition to differentiation. In the proliferative stage, immature SEPs and cells in the niche increased expression of inducible nitric oxide synthase (Nos2oriNOS) to generate NO. Increased NO rewires SEP metabolism to increase anabolic pathways, which drive the biosynthesis of nucleotides, amino acids and other intermediates needed for cell division. This NO-dependent metabolism promotes cell proliferation while also inhibiting erythroid differentiation leading to the amplification of a large population of non-committed progenitors. The transition of these progenitors to differentiation is mediated by the activation of nuclear factor erythroid 2-related factor 2 (Nfe2l2 or Nrf2). Nrf2 acts as an anti-inflammatory regulator that decreases NO production, which removes the NO-dependent erythroid inhibition and allows for differentiation. These data provide a paradigm for how alterations in metabolism allow inflammatory signals to amplify immature progenitors prior to differentiation.Key pointsNitric-oxide (NO) dependent signaling favors an anabolic metabolism that promotes proliferation and inhibits differentiation.Activation of Nfe2l2 (Nrf2) decreases NO production allowing erythroid differentiation.
- Published
- 2023
62. Multi-Omics Strategies for Investigating the Microbiome in Toxicology Research
- Author
-
Ethan W Morgan, Gary H Perdew, and Andrew D Patterson
- Subjects
Bacteria ,Microbiota ,Metabolomics ,Toxicology ,In-Depth Review ,Gastrointestinal Microbiome ,Xenobiotics - Abstract
Microbial communities on and within the host contact environmental pollutants, toxic compounds, and other xenobiotic compounds. These communities of bacteria, fungi, viruses, and archaea possess diverse metabolic potential to catabolize compounds and produce new metabolites. Microbes alter chemical disposition thus making the microbiome a natural subject of interest for toxicology. Sequencing and metabolomics technologies permit the study of microbiomes altered by acute or long-term exposure to xenobiotics. These investigations have already contributed to and are helping to re-interpret traditional understandings of toxicology. The purpose of this review is to provide a survey of the current methods used to characterize microbes within the context of toxicology. This will include discussion of commonly used techniques for conducting omic-based experiments, their respective strengths and deficiencies, and how forward-looking techniques may address present shortcomings. Finally, a perspective will be provided regarding common assumptions that currently impede microbiome studies from producing causal explanations of toxicologic mechanisms.
- Published
- 2022
63. Complex chemical signals dictate Ah receptor activation through the gut-lung axis
- Author
-
Fangcong Dong, Iain A. Murray, Andrew Annalora, Denise Coslo, Dhimant Desai, Krishne Gowda, Jian Yang, Dingbowen Wang, Imhoi Koo, Fuhua Hao, Shantu G. Amin, Andrew D. Patterson, Craig Marcus, and Gary H. Perdew
- Subjects
Article - Abstract
The aryl hydrocarbon receptor (AHR) mediates intestinal barrier homeostasis. Many AHR ligands are also CYP1A1/1B1 substrates, which can result in the rapid clearance within the intestinal tract, limiting AHR activation. This led us to the hypothesis that there are dietary substrates of CYP1A1/1B1 that increase the half-life of potent AHR ligands. We examined the potential of urolithin A (UroA) as a CYP1A1/1B1 substrate to enhance AHR activity in vivo. UroA is a competitive substrate for CYP1A1/1B1 in an in vitro competition assay. A broccoli-containing diet promotes the gastric formation of the potent hydrophobic AHR ligand and CYP1A1/1B1 substrate, 5,11-dihydroindolo[3,2-b]carbazole (ICZ). Dietary exposure to UroA in a broccoli diet led to a coordinated increase in duodenal, cardiac, and pulmonary AHR activity, but no increase in activity in liver. Thus, CYP1A1 dietary competitive substrates can lead to intestinal “escape”, likely through the lymphatic system, increasing AHR activation in key barrier tissues.
- Published
- 2023
64. Contribution of circulating host and microbial tryptophan metabolites towards Ah receptor activation
- Author
-
Ethan W. Morgan, Fangcong Dong, Andrew Annalora, Iain A. Murray, Trenton Wolfe, Reece Erickson, Krishne Gowda, Shantu G. Amin, Kristina S. Petersen, Penny M. Kris-Etherton, Craig Marcus, Seth T. Walk, Andrew D. Patterson, and Gary H. Perdew
- Subjects
Article - Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor that plays an integral role in homeostatic maintenance by regulating cellular functions such as cellular differentiation, metabolism, barrier function, and immune response. An important but poorly understood class of AHR activators are compounds derived from host and bacterial metabolism of tryptophan. The commensal bacteria of the gut microbiome are major producers of tryptophan metabolites known to activate the AHR, while the host also produces AHR activators through tryptophan metabolism. We used targeted mass spectrometry-based metabolite profiling to determine the presence and metabolic source of these metabolites in the sera of conventional mice, germ-free mice, and humans. Surprisingly, sera concentrations of many tryptophan metabolites are comparable between germ-free and conventional mice. Therefore, many major AHR-activating tryptophan metabolites in mouse sera are produced by the host, despite their presence in feces and mouse cecal contents. AHR activation is rarely studied in the context of a mixture at relevant concentrations, as we present here. The AHR activation potentials of individual and pooled metabolites were explored using cell-based assays, while ligand binding competition assays and ligand docking simulations were used to assess the detected metabolites as AHR agonists. The physiological and biomedical relevance of the identified metabolites was investigated in the context of cell-based models for cancer and rheumatoid arthritis. We present data here that reframe AHR biology to include the presence of ubiquitous tryptophan metabolites, improving our understanding of homeostatic AHR activity and models of AHR-linked diseases.
- Published
- 2023
65. The Underappreciated Diversity of Bile Acid Modifications
- Author
-
Ipsita Mohanty, Helena Mannochio-Russo, Yasin El Abiead, Joshua V. Schweer, Wout Bittremieux, Shipei Xing, Robin Schmid, Simone Zuffa, Felipe Vasquez, Valentina B. Muti, Jasmine Zemlin, Omar E. Tovar-Herrera, Sarah Moraïs, Dhimant Desai, Shantu Amin, Imhoi Koo, Christoph W. Turck, Itzhak Mizrahi, Tao Huan, Andrew D. Patterson, Dionicio Siegel, Lee R. Hagey, Mingxun Wang, Allegra T. Aron, and Pieter Dorrestein
- Published
- 2023
66. Loss of selenoprotein W in murine macrophages alters the hierarchy of selenoprotein expression, redox tone, and mitochondrial functions during inflammation
- Author
-
Sougat Misra, Tai-Jung Lee, Aswathy Sebastian, John McGuigan, Chang Liao, Imhoi Koo, Andrew D. Patterson, Randall M. Rossi, Molly A. Hall, Istvan Albert, and K. Sandeep Prabhu
- Subjects
Organic Chemistry ,Clinical Biochemistry ,Biochemistry - Abstract
Macrophages play a pivotal role in mediating inflammation and subsequent resolution of inflammation. The availability of selenium as a micronutrient and the subsequent biosynthesis of selenoproteins, containing the 21
- Published
- 2022
67. Current Challenges and Recent Developments in Mass Spectrometry–Based Metabolomics
- Author
-
Andrew D. Patterson, Jeffrey M. Peters, Philip B. Smith, Stephanie L. Collins, and Imhoi Koo
- Subjects
0301 basic medicine ,Databases, Factual ,Computer science ,Metabolite ,010401 analytical chemistry ,Computational biology ,Mass spectrometry ,01 natural sciences ,Mass Spectrometry ,0104 chemical sciences ,Analytical Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Chromatographic separation ,030104 developmental biology ,Metabolomics ,chemistry ,Mass spectrum ,Software ,Chromatography, Liquid - Abstract
High-resolution mass spectrometry (MS) has advanced the study of metabolism in living systems by allowing many metabolites to be measured in a single experiment. Although improvements in mass detector sensitivity have facilitated the detection of greater numbers of analytes, compound identification strategies, feature reduction software, and data sharing have not kept up with the influx of MS data. Here, we discuss the ongoing challenges with MS-based metabolomics, including de novo metabolite identification from mass spectra, differentiation of metabolites from environmental contamination, chromatographic separation of isomers, and incomplete MS databases. Because of their popularity and sensitive detection of small molecules, this review focuses on the challenges of liquid chromatography-mass spectrometry–based methods. We then highlight important instrumentational, experimental, and computational tools that have been created to address these challenges and how they have enabled the advancement of metabolomics research.
- Published
- 2021
68. Comparison of the effects of short-term feeding of sodium acetate and sodium bicarbonate on milk fat production
- Author
-
C. Matamoros, Jingwei Cai, Kevin J. Harvatine, and Andrew D. Patterson
- Subjects
Rumen ,Sodium Acetate ,Sodium ,chemistry.chemical_element ,Total mixed ration ,Article ,Milking ,chemistry.chemical_compound ,Animal science ,Genetics ,Animals ,Lactation ,Dry matter ,Sodium bicarbonate ,Fatty Acids ,Animal Feed ,Diet ,Neutral Detergent Fiber ,Milk ,Sodium Bicarbonate ,chemistry ,Dietary Supplements ,Lipogenesis ,Cattle ,Digestion ,Female ,Animal Science and Zoology ,Sodium acetate ,Food Science - Abstract
Supplementation with sodium acetate (NaAcet) increases milk fat production through an apparent stimulation of de novo lipogenesis in the mammary gland. Sodium acetate increases acetate supply to the mammary gland, but it also increases dietary cation-anion difference, which can also increase milk fat yield. The objective of this study was to determine if the effect of NaAcet on milk fat production was due to an increase in acetate supply or an increase in dietary cation-anion difference. The study included 12 multiparous cows in a replicated 3 × 3 Latin square design balanced for carryover effects, with 14-d experimental periods. Treatments were a basal total mixed ration (31.8% neutral detergent fiber, 14.8% crude protein, 25.5% starch, and 4.4% fatty acids on a dry matter basis) as a no-supplement control, acetate supplemented at 3.25% of dry matter as NaAcet, and sodium bicarbonate (NaHCO(3)) providing an equal amount of sodium to the NaAcet treatment. The NaAcet and NaHCO(3) were mixed into the basal diet before feeding. Milk samples were taken at each milking during the last 3 d of each period. Plasma samples were taken every 9 h during the last 3 d (a total of 8 times) to determine concentrations of plasma metabolites and hormones. Eating behavior was monitored during the last week of each period using an automated system. The NaAcet and NaHCO(3) treatments increased milk fat concentration and yield compared to the no-supplement control. The NaAcet treatment increased milk fat production predominantly by increasing the yield of de novo and mixed-source fatty acids. The NaHCO(3) treatment increased the yield of preformed and de novo fatty acids, suggesting different mechanisms for the 2 treatments. The NaAcet treatment increased plasma acetate concentration in a period of the day concurrent with the highest dry matter intake. The NaAcet treatment increased milk fat production by stimulating the production of de novo fatty acids, a mechanism consistent with previous reports, possibly by increasing acetate supply to the mammary gland. The NaHCO(3) treatment increased milk fat production by increasing the production of all biological categories of fatty acids, except for odd and branched-chain fatty acids, possibly by increasing overall diet digestibility.
- Published
- 2021
69. Caloric restriction disrupts the microbiota and colonization resistance
- Author
-
Marie Friedrich, Sophia Dickmann, Jingwei Cai, Jordan E. Bisanz, Danielle Ingebrigtsen, Katherine S. Pollard, Steve Miller, Knut Mai, Svetlana Lyalina, Peter J. Turnbaugh, Stephanie L. Collins, Qi Yan Ang, Reiner Jumpertz von Schwartzenberg, Andrew D. Patterson, Joachim Spranger, Peter Spanogiannopoulos, Jessie A. Turnbaugh, and Su Yang Liu
- Subjects
Male ,Diet, Reducing ,General Science & Technology ,Bacterial Toxins ,Calorie restriction ,Microbial metabolism ,Physiology ,Colonisation resistance ,Biology ,Article ,Oral and gastrointestinal ,Intestinal absorption ,Bile Acids and Salts ,Mice ,03 medical and health sciences ,Weight loss ,Weight Loss ,medicine ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Animals ,Humans ,Obesity ,Microbiome ,Aetiology ,Symbiosis ,Metabolic and endocrine ,Nutrition ,Adiposity ,Caloric Restriction ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Bacteria ,Clostridioides difficile ,030306 microbiology ,Prevention ,Reducing ,Body Weight ,Nutrients ,medicine.disease ,Diet ,Gastrointestinal Microbiome ,Transplantation ,Intestinal Absorption ,Metabolic syndrome ,medicine.symptom ,Digestive Diseases ,Energy Metabolism - Abstract
Diet is a major factor that shapes the gut microbiome1, but the consequences of diet-induced changes in the microbiome for host pathophysiology remain poorly understood. We conducted a randomized human intervention study using a very-low-calorie diet (NCT01105143). Although metabolic health was improved, severe calorie restriction led to a decrease in bacterial abundance and restructuring of the gut microbiome. Transplantation of post-diet microbiota to mice decreased their body weight and adiposity relative to mice that received pre-diet microbiota. Weight loss was associated with impaired nutrient absorption and enrichment in Clostridioides difficile, which was consistent with a decrease in bile acids and was sufficient to replicate metabolic phenotypes in mice in a toxin-dependent manner. These results emphasize the importance of diet-microbiome interactions in modulating host energy balance and the need to understand the role of diet in the interplay between pathogenic and beneficial symbionts.
- Published
- 2021
70. Genetic differences in nicotine sensitivity and metabolism in C57BL/6J and NOD/ShiLtJ mouse strains
- Author
-
Laurel R. Seemiller, Lisa R. Goldberg, Phillip B. Smith, Jason Dennis, Andrew D. Patterson, and Thomas J. Gould
- Subjects
Pharmacology ,Male ,Mice, Inbred C57BL ,Cellular and Molecular Neuroscience ,Mice ,Nicotine ,Mice, Inbred NOD ,Tandem Mass Spectrometry ,Animals ,Mice, Inbred Strains ,Tobacco Use Disorder ,Chromatography, Liquid - Abstract
Genetic background impacts sensitivity to nicotine's rewarding and aversive effects and metabolism, which influences susceptibility to nicotine addiction. This is important because sensitivity to nicotine influences susceptibility to nicotine addiction. Thus, understanding genetic contribution to nicotine sensitivity can aid in identifying risk factors for nicotine addiction. Genetic variability in addiction phenotypes can be modeled in rodent systems, and comparisons of nicotine sensitivity in inbred mice can identify contributing genetic substrates. Our laboratory has identified differences in nicotine sensitivity in male mice from two inbred mouse strains, C57BL/6J and NOD/ShiLtJ. We found that the NOD/ShiLtJ strain experienced greater nicotine-induced locomotor depression and hypothermia than the C57BL/6J strain. To investigate possible differences in nicotine metabolism between strains, subjects were treated with acute nicotine and serum and urine samples were analyzed using LC-MS/MS to quantify nicotine and metabolites. This analysis revealed that NOD/ShiLtJ mice had similar serum nicotine but lower cotinine and 3'-hydroxycotinine levels after nicotine treatment when compared to C57BL/6J mice. Possible genetic factors mediating strain differences were identified by surveying nicotine sensitivity- and metabolism-related genes within the Mouse Phenome Database SNP retrieval tool. Polymorphisms were found in 15 of the 26 examined gene sequences. Liver expression levels of nicotine metabolism-related genes (Cyp2a5, Cyp2a4, and Aox1) were measured using qPCR. NOD/ShiLtJ mice showed lower expression of Cyp2a5 and Cyp2a4 and greater expression of Aox1 in liver tissue. These data demonstrate complex differences in nicotine sensitivity and metabolism driven by genetic differences between C57BL/6J and NOD/ShiLtJ inbred mouse strains.
- Published
- 2022
71. A thermogenic fat-epithelium cell axis regulates intestinal disease tolerance
- Author
-
Christopher Bowman, Kristina N. Braverman, Kirthana Ganeshan, Yuan Tian, James R. Bayrer, Kevin Man, Peter J. Turnbaugh, Jordan E. Bisanz, Andrew D. Patterson, Ajay Chawla, Veronica Escalante, and Biao Wang
- Subjects
Male ,Azoxymethane ,Adrenergic ,Cell Communication ,Plant disease resistance ,Stimulus (physiology) ,Biology ,Mice ,Adipose Tissue, Brown ,Immunity ,Adipocytes ,Cell axis ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Disease Resistance ,Multidisciplinary ,Dextran Sulfate ,Enterobacteriaceae Infections ,Experimental colitis ,Epithelial Cells ,Thermogenesis ,Neoplasms, Experimental ,Metabolism ,Biological Sciences ,Colitis ,Epithelium ,Cell biology ,medicine.anatomical_structure ,Colonic Neoplasms ,Citrobacter rodentium ,Female - Abstract
Significance Disease tolerance is a host defense strategy that protects tissues from damage caused by pathogens or the immune system. While the importance of disease tolerance is well established in pathogenic infections, its role in tissue protection in noninfectious diseases remains less well understood. Here, we provide a framework for investigating the mechanisms of disease tolerance in experimental models of colitis. We find that the program of disease tolerance is preferentially expressed in thermoneutral mice, which protects them from injury-induced colitis and inflammation-induced colon cancer. The expression of intestinal disease tolerance is mediated by an unexpected cross-talk between thermogenic adipocytes and intestinal epithelial cells, suggesting that disease tolerance programs are metabolically expensive and dependent on the energetic state of the host.
- Published
- 2020
72. Bacterial colonization reprograms the neonatal gut metabolome
- Author
-
Elliot S. Friedman, Patricia A. DeRusso, Yuan Tian, Anumita Saha-Shah, Michal A. Elovitz, Josephine Ni, Andrea Kelly, Jingwei Cai, Debolina Sarkar, Lisa M. Mattei, Gary D. Wu, Mark Goulian, Casey E. Hofstaedter, Benjamin A. Garcia, Chunyu Zhao, Frederic D. Bushman, Eileen G Ford, Kyle Bittinger, Qing Liu, Chiraag Kulkarni, Peder J. Lund, Costas D. Maranas, Jeffrey S. Gerber, Dorothy Kim, Hongzhe Li, Siu Hung Joshua Chan, Andrew D. Patterson, Yun Li, and Babette S. Zemel
- Subjects
Microbiology (medical) ,Immunology ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Microbiology ,Communicable Diseases ,Enterococcus faecalis ,Article ,03 medical and health sciences ,Feces ,Meconium ,Genetics ,medicine ,Metabolome ,Humans ,Escherichia coli ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,Bacteria ,030306 microbiology ,Cell Biology ,biology.organism_classification ,Amino acid ,Gastrointestinal Microbiome ,chemistry ,Proteome ,Fermentation - Abstract
Initial microbial colonization and later succession in the gut of human infants are linked to health and disease later in life. The timing of the appearance of the first gut microbiome, and the consequences for the early life metabolome, are just starting to be defined. Here, we evaluated the gut microbiome, proteome and metabolome in 88 African-American newborns using faecal samples collected in the first few days of life. Gut bacteria became detectable using molecular methods by 16 h after birth. Detailed analysis of the three most common species, Escherichia coli, Enterococcus faecalis and Bacteroides vulgatus, did not suggest a genomic signature for neonatal gut colonization. The appearance of bacteria was associated with reduced abundance of approximately 50 human proteins, decreased levels of free amino acids and an increase in products of bacterial fermentation, including acetate and succinate. Using flux balance modelling and in vitro experiments, we provide evidence that fermentation of amino acids provides a mechanism for the initial growth of E. coli, the most common early colonizer, under anaerobic conditions. These results provide a deep characterization of the first microbes in the human gut and show how the biochemical environment is altered by their appearance. Using a multi-omics approach to analyse meconium and stool samples from babies during the first few days of life, the authors show that the gut is detectably colonized within 16 h of birth, with Escherichia coli dominating, and that this correlates with proteome and metabolome changes including the fermentation of amino acids.
- Published
- 2020
73. Profiling of the human intestinal microbiome and bile acids under physiologic conditions using an ingestible sampling device
- Author
-
Dari Shalon, Rebecca Neal Culver, Jessica A. Grembi, Jacob Folz, Peter Treit, Les Dethlefsen, Xiandong Meng, Eitan Yaffe, Sean Spencer, Handuo Shi, Andrés Aranda-Díaz, Andrew D. Patterson, George Triadafilopoulos, Susan P. Holmes, Matthias Mann, Oliver Fiehn, David A. Relman, and Kerwyn Casey Huang
- Abstract
The spatiotemporal structure of the human microbiome and metabolome reflects and determines regional intestinal physiology and may have implications for disease. Yet, we know little about the distribution of microbes and their products in the gut because of reliance on stool samples and limited access only to some regions of the gut using endoscopy in fasting or sedated individuals. To address these deficiencies, we developed and evaluated a safe, ingestible device that collects samples from multiple regions of the human intestinal tract during normal digestion. The collection of 240 intestinal samples from 15 healthy individuals using the device revealed significant differences between microbes and metabolites present in the intestines versus stool. Certain microbial taxa were differentially enriched, and bile acid profiles varied along the intestines and were highly distinct from those of stool. Correlations between gradients in bile acid concentrations and microbial abundance predicted species that altered the bile acid pool through deconjugation. Overall, we identified heterogeneous intestinal profiles of bacterial taxa and metabolites indicating that non-invasive multi-regional sampling of the intestinal tract under physiological conditions can help elucidate the roles of the gut microbiome and metabolome in human physiology and disease.
- Published
- 2022
74. Metabolomic profiling of stool of two-year old children from the INSIGHT study reveals links between butyrate and child weight outcomes
- Author
-
Francesca Chiaromonte, Matthew Reimherr, Ian M. Paul, Jingwei Cai, Sarah J. C. Craig, Michele E. Marini, Debmalya Nandy, Kateryna D. Makova, Andrew D. Patterson, Jennifer S. Savage, Emily E. Hohman, and Yuan Tian
- Subjects
1-HNMR ,Pediatric Obesity ,Maternal smoking ,Physiology ,Mothers ,Butyrate ,Gut flora ,Childhood obesity ,Body Mass Index ,Feces ,Pregnancy ,medicine ,Humans ,Maternal health ,Child ,Preschool ,Nutrition and Dietetics ,biology ,butyrate ,childhood obesity ,metabolomics ,weight outcomes at 2-years ,Butyrates ,Child, Preschool ,Female ,Gastrointestinal Microbiome ,business.industry ,Health Policy ,Public Health, Environmental and Occupational Health ,medicine.disease ,biology.organism_classification ,Obesity ,Metabolomic profiling ,Pediatrics, Perinatology and Child Health ,business - Abstract
Background Metabolomic analysis is commonly used to understand the biological underpinning of diseases such as obesity. However, our knowledge of gut metabolites related to weight outcomes in young children is currently limited. Objectives To (1) explore the relationships between metabolites and child weight outcomes, (2) determine the potential effect of covariates (e.g., child's diet, maternal health/habits during pregnancy, etc.) in the relationship between metabolites and child weight outcomes, and (3) explore the relationship between selected gut metabolites and gut microbiota abundance. Methods Using 1 H-NMR, we quantified 30 metabolites from stool samples of 170 two-year-old children. To identify metabolites and covariates associated with children's weight outcomes (BMI [weight/height2 ], BMI z-score [BMI adjusted for age and sex], and growth index [weight/height]), we analysed the 1 H-NMR data, along with 20 covariates recorded on children and mothers, using LASSO and best subset selection regression techniques. Previously characterized microbiota community information from the same stool samples was used to determine associations between selected gut metabolites and gut microbiota. Results At age 2 years, stool butyrate concentration had a significant positive association with child BMI (p-value = 3.58 × 10-4 ), BMI z-score (p-value = 3.47 × 10-4 ), and growth index (p-value = 7.73 × 10-4 ). Covariates such as maternal smoking during pregnancy are important to consider. Butyrate concentration was positively associated with the abundance of the bacterial genus Faecalibacterium (p-value = 9.61 × 10-3 ). Conclusions Stool butyrate concentration is positively associated with increased child weight outcomes and should be investigated further as a factor affecting childhood obesity.
- Published
- 2022
75. Aryl Hydrocarbon Receptor Activation Coordinates Mouse Small Intestinal Epithelial Cell Programming
- Author
-
Xiaoliang Zhou, Debopriya Chakraborty, Iain A. Murray, Denise Coslo, Zoe Kehs, Anitha Vijay, Carolyn Ton, Dhimant Desai, Shantu G. Amin, Andrew D. Patterson, and Gary H. Perdew
- Subjects
Cell Biology ,Molecular Biology ,Pathology and Forensic Medicine - Published
- 2023
76. Reconstitution of the host holobiont in germ-free born male rats acutely increases bone growth and affects marrow cellular content
- Author
-
Yuan Tian, Jiyoun Yeo, Sudipta Baroi, Xi Cheng, Beng San Yeoh, Andrew D. Patterson, Matam Vijay-Kumar, Beata Lecka-Czernik, Piu Saha, Bina Joe, Saroj Chakraborty, Rachel M. Golonka, Blair Mell, Piotr J. Czernik, and Ahmed A. Abokor
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Bone Marrow Cells ,Butyrate ,Biology ,Gut flora ,Bone tissue ,Bone Lengthening ,Muscle hypertrophy ,Rats, Sprague-Dawley ,Feces ,Chondrocytes ,Bone Density ,Osteogenesis ,Internal medicine ,RNA, Ribosomal, 16S ,Genetics ,medicine ,Adipocytes ,Coprophagia ,Animals ,Germ-Free Life ,Cell Proliferation ,Bone growth ,Bone Development ,Bacteria ,Host Microbial Interactions ,biology.organism_classification ,Fatty Acids, Volatile ,Gastrointestinal Microbiome ,Rats ,Endocrinology ,medicine.anatomical_structure ,Alkaline phosphatase ,Dysbiosis ,Bone marrow ,Research Article - Abstract
Integration of microbiota in a host begins at birth and progresses during adolescence, forming a multidirectional system of physiological interactions. Here, we present an instantaneous effect of natural, bacterial gut colonization on the acceleration of longitudinal and radial bone growth in germ-free born, 7-wk-old male rats. Changes in bone mass and structure were analyzed after 10 days following the onset of colonization through cohousing with conventional rats and revealed unprecedented acceleration of bone accrual in cortical and trabecular compartments, increased bone tissue mineral density, improved proliferation and hypertrophy of growth plate chondrocytes, bone lengthening, and preferential deposition of periosteal bone in the tibia diaphysis. In addition, the number of small in size adipocytes increased, whereas the number of megakaryocytes decreased, in the bone marrow of conventionalized germ-free rats indicating that not only bone mass but also bone marrow environment is under control of gut microbiota signaling. The changes in bone status paralleled with a positive shift in microbiota composition toward short-chain fatty acids (SCFA)-producing microbes and a considerable increase in cecal SCFA concentrations, specifically butyrate. Furthermore, reconstitution of the host holobiont increased hepatic expression of IGF-1 and its circulating levels. Elevated serum levels of 25-hydroxy vitamin D and alkaline phosphatase pointed toward an active process of bone formation. The acute stimulatory effect on bone growth occurred independently of body mass increase. Overall, the presented model of conventionalized germ-free rats could be used to study microbiota-based therapeutics for combatting dysbiosis-related bone disorders.
- Published
- 2021
77. Author response: The East Asian gut microbiome is distinct from colocalized White subjects and connected to metabolic health
- Author
-
Qi Yan Ang, Diana L. Alba, Vaibhav Upadhyay, Andrew D. Patterson, Peter J. Turnbaugh, Grace Wei, Eliseo Barajas, Jingwei Cai, Suneil K. Koliwad, Ho Lim Lee, Jordan E. Bisanz, and Cecilia Noecker
- Subjects
White (horse) ,Zoology ,East Asia ,Biology ,Gut microbiome ,Metabolic health - Published
- 2021
78. The East Asian gut microbiome is distinct from colocalized White subjects and connected to metabolic health
- Author
-
Eliseo Barajas, Suneil K. Koliwad, Peter J. Turnbaugh, Ho Lim Lee, Andrew D. Patterson, Qi Yan Ang, Jordan E. Bisanz, Jingwei Cai, Grace Wei, Vaibhav Upadhyay, Diana L. Alba, and Cecilia Noecker
- Subjects
obesity ,Far East ,Mouse ,Ethnic group ,Eastern ,Gut flora ,Oral and gastrointestinal ,California ,Feces ,2.1 Biological and endogenous factors ,Aetiology ,Biology (General) ,Microbiology and Infectious Disease ,biology ,Asia, Eastern ,General Neuroscience ,General Medicine ,Health equity ,Medicine ,ethnicity ,Akkermansia muciniphila ,Research Article ,Human ,Asia ,QH301-705.5 ,infectious disease ,1.1 Normal biological development and functioning ,Science ,Zoology ,Bacterial Physiological Phenomena ,General Biochemistry, Genetics and Molecular Biology ,metabolic syndrome ,Clinical Research ,Underpinning research ,Genetics ,medicine ,human ,Microbiome ,Life Below Water ,Metabolic and endocrine ,biogeography ,Nutrition ,human gut microbiome ,General Immunology and Microbiology ,Bacteria ,microbiology ,Human Genome ,multi-omics ,biology.organism_classification ,medicine.disease ,Obesity ,Gastrointestinal Microbiome ,Good Health and Well Being ,Metabolism ,Metagenomics ,Metagenome ,San Francisco ,Biochemistry and Cell Biology ,Metabolic syndrome - Abstract
East Asians (EAs) experience worse metabolic health outcomes compared to other ethnic groups at lower body mass indices; however, the potential role of the gut microbiota in contributing to these health disparities remains unknown. We conducted a multi-omic study of 46 lean and obese East Asian and White participants living in the San Francisco Bay Area, revealing marked differences between ethnic groups in bacterial richness and community structure. White individuals were enriched for the mucin-degrading Akkermansia muciniphila. East Asian subjects had increased levels of multiple bacterial phyla, fermentative pathways detected by metagenomics, and the short-chain fatty acid end-products acetate, propionate, and isobutyrate. Differences in the gut microbiota between the East Asian and White subjects could not be explained by dietary intake, were more pronounced in lean individuals, and were associated with current geographical location. Microbiome transplantations into germ-free mice demonstrated stable diet- and host genotype-independent differences between the gut microbiotas of East Asian and White individuals that differentially impact host body composition. Taken together, our findings add to the growing body of literature describing microbiome variations between ethnicities and provide a starting point for defining the mechanisms through which the microbiome may shape disparate health outcomes in East Asians., eLife digest The community of microbes living in the human gut varies based on where a person lives, in part because of differences in diets but also due to factors still incompletely understood. In turn, this ‘microbiome’ may have wide-ranging effects on health and diseases such as obesity and diabetes. Many scientists want to understand how differences in the microbiome emerge between people, and whether this may explain why certain diseases are more common in specific populations. Self-identified race or ethnicity can be a useful tool in that effort, as it can serve as a proxy for cultural habits (such as diets) or genetic information. In the United States, self-identified East Asian Americans often have worse ‘metabolic health’ (e.g. levels of sugar or certain fat molecules in the blood) at a lower weight than those identifying as White. Ang, Alba, Upadhyay et al. investigated whether this health disparity was linked to variation in the gut microbiome. Samples were collected from 46 lean and obese individuals living in the San Francisco Bay Area who identified as White or East Asian. The analyses showed that while the gut microbiome of White participants changed in association with obesity, the microbiomes of East Asian participants were distinct from their White counterparts even at normal weight, with features mirroring what was seen in White individuals in the context of obesity. Although these differences were connected to people’s current address, they were not attributable to dietary differences. Ang, Alba, Upadhyay et al. then transplanted the microbiome of the participants into genetically identical mice with microbe-free guts. The differences between the gut microbiomes of White and East Asian participants persisted in recipient animals. When fed the same diet, the mice also gained different amounts of weight depending on the ethnic identity of the microbial donor. These results show that self-identified ethnicity may be an important variable to consider in microbiome studies, alongside other factors such as geography. Ultimately, this research may help to design better, more personalized treatments for an array of conditions.
- Published
- 2021
79. The role of mouse and human peroxisome proliferator-activated receptor-α in modulating the hepatic effects of perfluorooctane sulfonate in mice
- Author
-
Shengzhong Su, Jeffrey M. Peters, Frank J. Gonzalez, Sue Chang, Andrew D. Patterson, and Laura J. Billy
- Subjects
Male ,medicine.medical_specialty ,Mice, 129 Strain ,Peroxisome Proliferation ,Peroxisome proliferator-activated receptor ,Toxicology ,Article ,chemistry.chemical_compound ,Cytochrome P-450 Enzyme System ,Species Specificity ,Internal medicine ,Constitutive androstane receptor ,medicine ,Animals ,Humans ,PPAR alpha ,Receptor ,Cytochrome P450 Family 2 ,Constitutive Androstane Receptor ,chemistry.chemical_classification ,Mice, Knockout ,Pregnane X receptor ,Fluorocarbons ,Dose-Response Relationship, Drug ,Chemistry ,Wild type ,Pregnane X Receptor ,Perfluorooctane ,Endocrinology ,Alkanesulfonic Acids ,Liver ,Steroid Hydroxylases ,ACOX1 ,Environmental Pollutants ,Acyl-CoA Oxidase ,Aryl Hydrocarbon Hydroxylases ,Chemical and Drug Induced Liver Injury ,Signal Transduction - Abstract
Perfluorooctane sulfonate (PFOS) is a stable environmental contaminant that can activate peroxisome proliferator-activated receptor alpha (PPARα). In the present work, the specific role of mouse and human PPARα in mediating the hepatic effects of PFOS was examined in short-term studies using wild type, Ppara-null and PPARA-humanized mice. Mice fed 0.006 % PFOS for seven days (~10 mg/kg/day), or 0.003 % PFOS for twenty-eight days (~5 mg/kg/day), exhibited higher liver and serum PFOS concentrations compared to controls. Relative liver weights were also higher following exposure to dietary PFOS in all three genotypes as compared vehicle fed control groups. Histopathological examination of liver sections from mice treated for twenty-eight days with 0.003 % PFOS revealed a phenotype consistent with peroxisome proliferation, in wild-type and PPARA-humanized mice that was not observed in Ppara-null mice. With both exposures, expression of the PPARα target genes, Acox1, Cyp4a10, was significantly increased in wild type mice but not in Ppara-null or PPARA-humanized mice. By contrast, expression of the constitutive androstane receptor (CAR) target gene, Cyp2b10, and the pregnane × receptor (PXR) target gene, Cyp3a11, were higher in response to PFOS administration in all three genotypes compared to controls for both exposure periods. These results indicate that mouse PPARα can be activated in the liver by PFOS causing increased expression of Acox1, Cyp4a10 and histopathological changes in the liver. While histopathological analyses indicated the presence of mouse PPARα-dependent hepatic peroxi-some proliferation in wild-type (a response associated with activation of PPARα) and a similar phenotype in PPARA-humanized mice, the lack of increased Acox1 and Cyp4a10 mRNA by PFOS in PPARA-humanized mice indicates that the human PPARα was not as responsive to PFOS as mouse PPARα with this dose regimen. Moreover, results indicate that hepatomegaly caused by PFOS does not require mouse or human PPARα and could be due to effects induced by activation of CAR and/or PXR.
- Published
- 2021
80. Bile acid metabolism and signaling, the microbiota, and metabolic disease
- Author
-
Jingwei Cai, Bipin Rimal, Changtao Jiang, John Y.L. Chiang, and Andrew D. Patterson
- Subjects
Bile Acids and Salts ,Pharmacology ,Bacteria ,Metabolic Diseases ,Microbiota ,Humans ,Pharmacology (medical) ,Lipid Metabolism ,Gastrointestinal Microbiome - Abstract
The diversity, composition, and function of the bacterial community inhabiting the human gastrointestinal tract contributes to host health through its role in producing energy or signaling molecules that regulate metabolic and immunologic functions. Bile acids are potent metabolic and immune signaling molecules synthesized from cholesterol in the liver and then transported to the intestine where they can undergo metabolism by gut bacteria. The combination of host- and microbiota-derived enzymatic activities contribute to the composition of the bile acid pool and thus there can be great diversity in bile acid composition that depends in part on the differences in the gut bacteria species. Bile acids can profoundly impact host metabolic and immunological functions by activating different bile acid receptors to regulate signaling pathways that control a broad range of complex symbiotic metabolic networks, including glucose, lipid, steroid and xenobiotic metabolism, and modulation of energy homeostasis. Disruption of bile acid signaling due to perturbation of the gut microbiota or dysregulation of the gut microbiota-host interaction is associated with the pathogenesis and progression of metabolic disorders. The metabolic and immunological roles of bile acids in human health have led to novel therapeutic approaches to manipulate the bile acid pool size, composition, and function by targeting one or multiple components of the microbiota-bile acid-bile acid receptor axis.
- Published
- 2022
81. A Synthesis-Based Reverse Metabolomics Approach for the Discovery of Chemical Structures from Humans and Animals
- Author
-
Andrew D. Patterson, Stephanie L. Collins, Ramnik J. Xavier, Clary B. Clish, Robin Knight, Emily C. Gentry, Pieter C. Dorrestein, Erin S. Baker, Hera Vlamakis, Allegra T. Aron, Damian R. Plichta, Pedro Belda-Ferre, Alan K. Jarmusch, Morgan Panitchpakdi, Ashwin N. Ananthakrishnan, Dionicio Siegel, Mingxun Wang, Allison Stewart, and Julian Avila-Pacheco
- Subjects
Metabolomics ,Computational biology ,Biology - Abstract
Identification of metabolites in humans remains challenging. Here, we present synthesis-based reverse metabolomics as a strategy for structure elucidation that aims to also find phenotypic associations. In this approach, MS/MS spectra are acquired from newly synthesized compounds, and searched against public metabolomics data to uncover their phenotypic associations. To demonstrate the concept, we used combinatorial amide coupling reactions to synthesize an array of amino acid conjugated bile acids. A total of 16,587 spectral matches were found for 145 amidates synthesized, representing the single largest expansion of structures in the 170+ year history of bile acids. Furthermore, some new bile acids were associated with health-related phenotypes such as inflammatory bowel diseases and obesity. Using independent human cohorts for validation revealed that some cholic and chenodeoxycholic acid conjugates were elevated in Crohn’s disease, of which some were also potent PXR agonists. Bacteria belonging to bifidobacterium, clostridium, and enterococci genera were the main producers of these new conjugated bile acids. Because searching repositories with MS/MS spectra has only recently become possible, this synthesis-based reverse metabolomics approach can now be employed as a general strategy to elucidate structures and discover other new molecules from human and animal ecosystems.
- Published
- 2021
82. Exercise Training Reverses Gut Dysbiosis in Patients With Biopsy-Proven Nonalcoholic Steatohepatitis: A Proof of Concept Study
- Author
-
Jessica Dahmus, Regina Lamendella, Gloriany Rivas, Justin P. Wright, Christopher N. Sciamanna, Rohit Loomba, Jeremy R. Chen See, Kathryn H. Schmitz, Jonathan G. Stine, Breianna L. Hummer, Andrew D. Patterson, Alexa Hughes, and Mack T. Ruffin
- Subjects
Nonalcoholic steatohepatitis ,medicine.medical_specialty ,Biopsy ,Chronic Liver Disease and Cirrhosis ,Clinical Sciences ,digestive system ,Gastroenterology ,Proof of Concept Study ,Article ,Oral and gastrointestinal ,Hepatitis ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Clinical Research ,Internal medicine ,Nonalcoholic fatty liver disease ,medicine ,Animals ,Humans ,In patient ,Exercise ,Hepatology ,medicine.diagnostic_test ,Gastroenterology & Hepatology ,business.industry ,Prevention ,Liver Disease ,nutritional and metabolic diseases ,medicine.disease ,Obesity ,digestive system diseases ,Gastrointestinal Microbiome ,Good Health and Well Being ,Liver ,030220 oncology & carcinogenesis ,Dysbiosis ,030211 gastroenterology & hepatology ,Gut dysbiosis ,business ,Digestive Diseases - Abstract
Nonalcoholic fatty liver disease is the leading cause of liver disease worldwide and can progress to nonalcoholic steatohepatitis (NASH) through physical inactivity and gut dysbiosis.1 Exercise training reverses gut dysbiosis in non-NASH persons with obesity and in NASH animal models.2,3 Consequently, we conducted a proof-of-concept study investigating the effect of exercise training on gut dysbiosis in NASH patients.
- Published
- 2021
83. A microbiome-dependent gut-brain pathway regulates motivation for exercise
- Author
-
Lenka, Dohnalová, Patrick, Lundgren, Jamie R E, Carty, Nitsan, Goldstein, Sebastian L, Wenski, Pakjira, Nanudorn, Sirinthra, Thiengmag, Kuei-Pin, Huang, Lev, Litichevskiy, Hélène C, Descamps, Karthikeyani, Chellappa, Ana, Glassman, Susanne, Kessler, Jihee, Kim, Timothy O, Cox, Oxana, Dmitrieva-Posocco, Andrea C, Wong, Erik L, Allman, Soumita, Ghosh, Nitika, Sharma, Kasturi, Sengupta, Belinda, Cornes, Nitai, Dean, Gary A, Churchill, Tejvir S, Khurana, Mark A, Sellmyer, Garret A, FitzGerald, Andrew D, Patterson, Joseph A, Baur, Amber L, Alhadeff, Eric J N, Helfrich, Maayan, Levy, J Nicholas, Betley, and Christoph A, Thaiss
- Abstract
Exercise exerts a wide range of beneficial effects for healthy physiology
- Published
- 2021
84. Unraveling the role of peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) expression in colon carcinogenesis
- Author
-
Jeffrey M. Peters, Vonn Walter, Frank J. Gonzalez, and Andrew D. Patterson
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Cancer Research ,Cell growth ,Peroxisome proliferator-activated receptor ,Endogeny ,Review Article ,Peroxisome ,medicine.disease_cause ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Colon cancer ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,Transcription (biology) ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,Receptor ,Carcinogenesis ,Cancer - Abstract
The peroxisome proliferator-activated-β/δ (PPARβ/δ) was identified in 1994, but not until 1999 was PPARβ/δ suggested to be involved in carcinogenesis. Initially, it was hypothesized that expression of PPARβ/δ was increased during colon cancer progression, which led to increased transcription of yet-to-be confirmed target genes that promote cell proliferation and tumorigenesis. It was also hypothesized at this time that lipid-metabolizing enzymes generated lipid metabolites that served as ligands for PPARβ/δ. These hypothetical mechanisms were attractive because they potentially explained how non-steroidal anti-inflammatory drugs inhibited tumorigenesis by potentially limiting the concentration of endogenous PPARβ/δ ligands that could activate this receptor that was increased in cancer cells. However, during the last 20 years, considerable research was undertaken describing expression of PPARβ/δ in normal and cancer cells that has led to a significant impact on the mechanisms by which PPARβ/δ functions in carcinogenesis. Whereas results from earlier studies led to much uncertainty about the role of PPARβ/δ in cancer, more recent analyses of large databases have revealed a more consistent understanding. The focus of this review is on the fundamental level of PPARβ/δ expression in normal tissues and cancerous tissue as described by studies during the past two decades and what has been delineated during this timeframe about how PPARβ/δ expression influences carcinogenesis, with an emphasis on colon cancer.
- Published
- 2019
85. Gdf15 regulates murine stress erythroid progenitor proliferation and the development of the stress erythropoiesis niche
- Author
-
Dai Chen Wu, Andrew D. Patterson, James Fraser, Siyang Hao, Zhi Chun Lai, Jingwei Cai, Robert F. Paulson, Jie Xiang, and Baiye Ruan
- Subjects
0301 basic medicine ,Growth Differentiation Factor 15 ,Cellular differentiation ,Regulator ,Spleen ,Biology ,Models, Biological ,Mice ,03 medical and health sciences ,Red Cells, Iron, and Erythropoiesis ,0302 clinical medicine ,Stress, Physiological ,hemic and lymphatic diseases ,medicine ,Animals ,Erythropoiesis ,Stem Cell Niche ,Progenitor cell ,Cell Proliferation ,Erythroid Precursor Cells ,Mice, Knockout ,Cell growth ,Cell Differentiation ,Hematology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Signal transduction ,Homeostasis ,Signal Transduction - Abstract
Anemic stress induces the proliferation of stress erythroid progenitors in the murine spleen that subsequently differentiate to generate erythrocytes to maintain homeostasis. This process relies on the interaction between stress erythroid progenitors and the signals generated in the splenic erythroid niche. In this study, we demonstrate that although growth-differentiation factor 15 (Gdf15) is not required for steady-state erythropoiesis, it plays an essential role in stress erythropoiesis. Gdf15 acts at 2 levels. In the splenic niche, Gdf15−/− mice exhibit defects in the monocyte-derived expansion of the splenic niche, resulting in impaired proliferation of stress erythroid progenitors and production of stress burst forming unit-erythroid cells. Furthermore, Gdf15 signaling maintains the hypoxia-dependent expression of the niche signal, Bmp4, whereas in stress erythroid progenitors, Gdf15 signaling regulates the expression of metabolic enzymes, which contribute to the rapid proliferation of stress erythroid progenitors. Thus, Gdf15 functions as a comprehensive regulator that coordinates the stress erythroid microenvironment with the metabolic status of progenitors to promote stress erythropoiesis.
- Published
- 2019
86. Interplay Between the Host, the Human Microbiome, and Drug Metabolism
- Author
-
Jeffrey M. Peters, Andrew D. Patterson, and Robert G. Nichols
- Subjects
lcsh:QH426-470 ,lcsh:Medicine ,Cytochrome P450 ,Review ,Biology ,Human microbiome ,Vaginal microbiome ,Dermatitis, Atopic ,Irritable Bowel Syndrome ,03 medical and health sciences ,Detoxification ,Drug Discovery ,Genetics ,medicine ,Humans ,Microbiome ,Molecular Biology ,Irritable bowel syndrome ,Drug metabolism ,0303 health sciences ,Microbiota ,Probiotics ,030305 genetics & heredity ,lcsh:R ,Vaginosis, Bacterial ,Atopic dermatitis ,medicine.disease ,Oral microbiome ,Gingivitis ,Human genetics ,Intestinal microbiome ,3. Good health ,lcsh:Genetics ,Prebiotics ,Inactivation, Metabolic ,Immunology ,Skin microbiome ,Molecular Medicine ,Female ,Oral Microbiome - Abstract
The human microbiome is composed of four major areas including intestinal, skin, vaginal, and oral microbiomes, with each area containing unique species and unique functionalities. The human microbiome may be modulated with prebiotics, probiotics, and postbiotics to potentially aid in the treatment of diseases like irritable bowel syndrome, bacterial vaginosis, atopic dermatitis, gingivitis, obesity, or cancer. There is also potential for many of the inhabitants of the human microbiome to directly modulate host gene expression and modulate host detoxifying enzyme activity like cytochrome P450s (CYPs), dehydrogenases, and carboxylesterases. Therefore, the microbiome may be important to consider during drug discovery, risk assessment, and dosing regimens for various diseases given that the human microbiome has been shown to impact host detoxification processes.
- Published
- 2019
87. Microbiota Metabolism Promotes Synthesis of the Human Ah Receptor Agonist 2,8-Dihydroxyquinoline
- Author
-
Troy D. Hubbard, Krishne Gowda, Jyh Ming Lin, Iain A. Murray, Qing Liu, Shantu Amin, Fangcong Dong, Philip B. Smith, Andrew D. Patterson, Gary H. Perdew, and Charles A. Miller
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,Regulator ,Biochemistry ,Article ,Feces ,Mice ,03 medical and health sciences ,Immune system ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Humans ,Gastrointestinal tract ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Tryptophan ,General Chemistry ,Metabolism ,respiratory system ,Oxyquinoline ,Aryl hydrocarbon receptor ,Gastrointestinal Microbiome ,respiratory tract diseases ,Cell biology ,030104 developmental biology ,Receptors, Aryl Hydrocarbon ,Caco-2 ,biology.protein ,Caco-2 Cells ,Signal transduction - Abstract
The aryl hydrocarbon receptor (AHR) is a major regulator of immune function within the gastrointestinal tract. Resident microbiota are capable of influencing AHR-dependent signaling pathways via production of an array of bioactive molecules that act as AHR agonists, such as indole or indole-3-aldehyde. Bacteria produce a number of quinoline derivatives, of which some function as quorum-sensing molecules. Thus, we screened relevant hydroxyquinoline derivatives for AHR activity using AHR responsive reporter cell lines. 2,8-Dihydroxyquinoline (2,8-DHQ) was identified as a species-specific AHR agonist that exhibits full AHR agonist activity in human cell lines, but only induces modest AHR activity in mouse cells. Additional dihydroxylated quinolines tested failed to activate the human AHR. Nanomolar concentrations of 2,8-DHQ significantly induced CYP1A1 expression and, upon cotreatment with cytokines, synergistically induced IL6 expression. Ligand binding competition studies subsequently confirmed 2,8-DHQ to be a human AHR ligand. Several dihydroxyquinolines were detected in human fecal samples, with concentrations of 2,8-DHQ ranging between 0 and 3.4 pmol/mg feces. Additionally, in mice the microbiota was necessary for the presence of DHQ in cecal contents. These results suggest that microbiota-derived 2,8-DHQ would contribute to AHR activation in the human gut, and thus participate in the protective and homeostatic effects observed with gastrointestinal AHR activation.
- Published
- 2019
88. Randomized Controlled-Feeding Study of Dietary Emulsifier Carboxymethylcellulose Reveals Detrimental Impacts on the Gut Microbiota and Metabolome
- Author
-
Benoit, Chassaing, Charlene, Compher, Brittaney, Bonhomme, Qing, Liu, Yuan, Tian, William, Walters, Lisa, Nessel, Clara, Delaroque, Fuhua, Hao, Victoria, Gershuni, Lillian, Chau, Josephine, Ni, Meenakshi, Bewtra, Lindsey, Albenberg, Alexis, Bretin, Liam, McKeever, Ruth E, Ley, Andrew D, Patterson, Gary D, Wu, Andrew T, Gewirtz, and James D, Lewis
- Subjects
Male ,Healthy Volunteers ,Diet ,Gastrointestinal Microbiome ,Feces ,Mice ,Double-Blind Method ,Carboxymethylcellulose Sodium ,Emulsifying Agents ,Metabolome ,Animals ,Dysbiosis ,Humans ,Female - Abstract
Epidemiologic and murine studies suggest that dietary emulsifiers promote development of diseases associated with microbiota dysbiosis. Although the detrimental impact of these compounds on the intestinal microbiota and intestinal health have been demonstrated in animal and in vitro models, impact of these food additives in healthy humans remains poorly characterized.To examine this notion in humans, we performed a double-blind controlled-feeding study of the ubiquitous synthetic emulsifier carboxymethylcellulose (CMC) in which healthy adults consumed only emulsifier-free diets (n = 9) or an identical diet enriched with 15 g per day of CMC (n = 7) for 11 days.Relative to control subjects, CMC consumption modestly increased postprandial abdominal discomfort and perturbed gut microbiota composition in a way that reduced its diversity. Moreover, CMC-fed subjects exhibited changes in the fecal metabolome, particularly reductions in short-chain fatty acids and free amino acids. Furthermore, we identified 2 subjects consuming CMC who exhibited increased microbiota encroachment into the normally sterile inner mucus layer, a central feature of gut inflammation, as well as stark alterations in microbiota composition.These results support the notion that the broad use of CMC in processed foods may be contributing to increased prevalence of an array of chronic inflammatory diseases by altering the gut microbiome and metabolome (ClinicalTrials.gov, number NCT03440229).
- Published
- 2021
89. Gut Microbiota Accelerates Bone Growth and Marrow Adiposity of the Adolescent Gnotobiotic Rat
- Author
-
Xi Cheng, Piotr J. Czernik, Matam Vijay-Kumar, Ahmed A. Abokor, Saroj Chakraborty, Jiyoun Yeo, Yuan Tian, Piu Saha, Bina Joe, Sudipta Baroi, Beata Lecka-Czernik, Beng San Yeoh, Andrew D. Patterson, Rachel M. Golonka, and Blair Mell
- Subjects
Bone growth ,medicine.medical_specialty ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Biology ,Gut flora ,biology.organism_classification ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2021
90. I ncreased Host Energy Metabolism in the Proximal Colon‐Microbiota Interface Elevates Blood Pressure
- Author
-
Matam Vijay-Kumar, Andrew D. Patterson, Jiyoun Yeo, Xi Cheng, Xue Mei, Yuan Tian, Rachel M. Golonka, Blair Mell, Tao Yang, Saroj Chakraborty, Piu Saha, and Bina Joe
- Subjects
Blood pressure ,Host (biology) ,Chemistry ,Genetics ,Energy metabolism ,Proximal colon ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2021
91. Metabolomics reveal dynamic host responses in lipid, amino acid, and energy metabolism after acute exposure of gut microbiota in germ‐free rats
- Author
-
Matam Vijay-Kumar, Beng San Yeoh, Piu Saha, Bina Joe, Xi Cheng, Rachel M. Golonka, Yuan Tian, Andrew D. Patterson, Blair Mell, Jiyoun Yeo, Ahmed A. Abokor, and Saroj Chakraborty
- Subjects
chemistry.chemical_classification ,Host (biology) ,Energy metabolism ,Biology ,Gut flora ,biology.organism_classification ,Biochemistry ,Amino acid ,Metabolomics ,chemistry ,Acute exposure ,Genetics ,Germ ,Molecular Biology ,Biotechnology - Published
- 2021
92. Role of bile acids and gut bacteria in healthy ageing of centenarians
- Author
-
Andrew D. Patterson and Bipin Rimal
- Subjects
Multidisciplinary ,biology ,Gut bacteria ,Healthy ageing ,biology.organism_classification ,Bacteria ,Microbiology - Abstract
A study in humans indicates that certain bile acids that are produced by bacteria and commonly found in people over 100 boost gut health and protect against infection. These findings shed light on the contributors to healthy ageing. Analysis points to factors that contribute to healthy ageing of humans.
- Published
- 2021
93. Impaired Intestinal <named-content content-type='genus-species'>Akkermansia muciniphila</named-content> and Aryl Hydrocarbon Receptor Ligands Contribute to Nonalcoholic Fatty Liver Disease in Mice
- Author
-
Manyuan Dong, Zunji Shi, Chuan Chen, Gui Chen, Hooi-Leng Ser, Limin Zhang, Xuehang Zhu, Peihong Yuan, Fang Wu, Yuchen Song, Caixiang Liu, Xin An Zeng, Yangyang Guo, Yifan Zhu, Zheng Cao, Yu-Jing Lu, Kexin Hu, Andrew D. Patterson, Jinlin Zhou, and Hehua Lei
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Sucralose ,Physiology ,microbiome ,Gut flora ,Biochemistry ,digestive system ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Nonalcoholic fatty liver disease ,Genetics ,medicine ,Molecular Biology ,Saccharin ,Ecology, Evolution, Behavior and Systematics ,Intestinal permeability ,gut-liver axis ,biology ,Chemistry ,digestive, oral, and skin physiology ,biology.organism_classification ,medicine.disease ,Aryl hydrocarbon receptor ,Editor's Pick ,QR1-502 ,Computer Science Applications ,Metformin ,AHR ligands ,030104 developmental biology ,Endocrinology ,030220 oncology & carcinogenesis ,Modeling and Simulation ,biology.protein ,Akkermansia muciniphila ,medicine.drug ,Research Article - Abstract
Noncaloric artificial sweeteners (NAS) are extensively introduced into commonly consumed drinks and foods worldwide. However, data on the health effects of NAS consumption remain elusive. Saccharin and sucralose have been shown to pass through the human gastrointestinal tract without undergoing absorption and metabolism and directly encounter the gut microbiota community. Here, we aimed to identify a novel mechanism linking intestinal Akkermansia muciniphila and the aryl hydrocarbon receptor (AHR) to saccharin/sucralose-induced nonalcoholic fatty liver disease (NAFLD) in mice. Saccharin/sucralose consumption altered the gut microbial community structure, with significant depletion of A. muciniphila abundance in the cecal contents of mice, resulting in disruption of intestinal permeability and a high level of serum lipopolysaccharide, which likely contributed to systemic inflammation and caused NAFLD in mice. Saccharin/sucralose also markedly decreased microbiota-derived AHR ligands and colonic AHR expression, which are closely associated with many metabolic syndromes. Metformin or fructo-oligosaccharide supplementation significantly restored A. muciniphila and AHR ligands in sucralose-consuming mice, consequently ameliorating NAFLD. IMPORTANCE Our findings indicate that the gut-liver signaling axis contributes to saccharin/sucralose consumption-induced NAFLD. Supplementation with metformin or fructo-oligosaccharide is a potential therapeutic strategy for NAFLD treatment. In addition, we also developed a new nutritional strategy by using a natural sweetener (neohesperidin dihydrochalcone [NHDC]) as a substitute for NAS and free sugars.
- Published
- 2021
94. Selenium-dependent metabolic reprogramming during inflammation and resolution
- Author
-
Philip B. Smith, Arvind M. Korwar, Ayaan Hossain, Tai Jung Lee, Bradley A. Carlson, Ashley E. Shay, Venkatesha Basrur, Howard M. Salis, K. Sandeep Prabhu, Andrew D. Patterson, and Kevin P. Conlon
- Subjects
Lipopolysaccharides ,Male ,Proteomics ,0301 basic medicine ,Proteome ,OXPHOS, oxidative phosphorylation ,Biochemistry ,Mice ,chemistry.chemical_compound ,Se, Selenium ,Selenoproteins ,Ri, resolution index ,Selenocysteine ,Chemistry ,Cell biology ,redox ,LPS, lipopolysaccharide ,Hif-1α, hypoxia inducible factor-1α ,Disease Susceptibility ,Sedoheptulokinase ,Research Article ,Sdh, succinate dehydrogenase complex ,TCA, tricarboxylic acid ,PPP, pentose phosphate pathway ,Mice, Transgenic ,Oxidative phosphorylation ,Peritonitis ,Pentose phosphate pathway ,PKM2 ,Selenium ,03 medical and health sciences ,ROS, reactive oxygen species ,Metabolomics ,Metabolome ,Animals ,Idh1, isocitrate dehydrogenase ,Pkm2, pyruvate kinase 2 ,Molecular Biology ,DMM, dimethylmalonate ,Carkl, carbohydrate-like kinase ,Inflammation ,Shpk, Sedoheptulokinase ,030102 biochemistry & molecular biology ,Macrophages ,TMT, tandem mass tag ,Cell Biology ,succinate dehydrogenase ,Mice, Inbred C57BL ,Citric acid cycle ,Metabolic pathway ,030104 developmental biology ,IL-4, interleukin-4 ,Trsp, tRNA[Ser]Sec ,Pyruvate kinase ,BMDMs, bone marrow–derived macrophages - Abstract
Trace element selenium (Se) is incorporated as the 21st amino acid, selenocysteine, into selenoproteins through tRNA[Ser]Sec. Selenoproteins act as gatekeepers of redox homeostasis and modulate immune function to effect anti-inflammation and resolution. However, mechanistic underpinnings involving metabolic reprogramming during inflammation and resolution remain poorly understood. Bacterial endotoxin lipopolysaccharide (LPS) activation of murine bone marrow–derived macrophages cultured in the presence or absence of Se (as selenite) was used to examine temporal changes in the proteome and metabolome by multiplexed tandem mass tag–quantitative proteomics, metabolomics, and machine-learning approaches. Kinetic deltagram and clustering analysis indicated that addition of Se led to extensive reprogramming of cellular metabolism upon stimulation with LPS enhancing the pentose phosphate pathway, tricarboxylic acid cycle, and oxidative phosphorylation, to aid in the phenotypic transition toward alternatively activated macrophages, synonymous with resolution of inflammation. Remodeling of metabolic pathways and consequent metabolic adaptation toward proresolving phenotypes began with Se treatment at 0 h and became most prominent around 8 h after LPS stimulation that included succinate dehydrogenase complex, pyruvate kinase, and sedoheptulokinase. Se-dependent modulation of these pathways predisposed bone marrow–derived macrophages to preferentially increase oxidative phosphorylation to efficiently regulate inflammation and its timely resolution. The use of macrophages lacking selenoproteins indicated that all three metabolic nodes were sensitive to selenoproteome expression. Furthermore, inhibition of succinate dehydrogenase complex with dimethylmalonate affected the proresolving effects of Se by increasing the resolution interval in a murine peritonitis model. In summary, our studies provide novel insights into the role of cellular Se via metabolic reprograming to facilitate anti-inflammation and proresolution.
- Published
- 2021
95. Tu1329: TCDD INDUCED AHR SIGNALING ALTERS ENTERIC NEURONAL MARKER EXPRESSION AND DELAYED INTESTINAL TRANSIT
- Author
-
Anitha Vijay, Shanthi Srinivasan, and Andrew D. Patterson
- Subjects
Hepatology ,Gastroenterology - Published
- 2022
96. The Pretreatment Gut Microbiome Is Associated With Lack of Response to Methotrexate in New-Onset Rheumatoid Arthritis
- Author
-
Antonio Pineda-Lucena, Renuka R. Nayak, Julia Manasson, Alejandra Flor-Duro, Pércio S. Gulko, Carles Ubeda, Yamen Homsi, Pamela Rosenthal, Imhoi Koo, Jose U. Scher, Alejandro Artacho, Margaret Alexander, Peter J. Turnbaugh, Leonor Puchades-Carrasco, Steven B. Abramson, Sandrine Isaac, Andrew D. Patterson, Javier Pons, Philip B. Smith, and Peter M. Izmirly
- Subjects
0301 basic medicine ,Purine ,Drug ,Adult ,Escherichia ,Male ,media_common.quotation_subject ,Immunology ,Administration, Oral ,Firmicutes ,Euryarchaeota ,Arthritis, Rheumatoid ,Cohort Studies ,Machine Learning ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Metabolomics ,Rheumatology ,RNA, Ribosomal, 16S ,medicine ,Immunology and Allergy ,Humans ,Microbiome ,media_common ,030203 arthritis & rheumatology ,Clostridiales ,business.industry ,Bacteroidetes ,Middle Aged ,medicine.disease ,Prognosis ,Gastrointestinal Microbiome ,030104 developmental biology ,Methotrexate ,Treatment Outcome ,chemistry ,Metagenomics ,Rheumatoid arthritis ,Antirheumatic Agents ,Female ,Shigella ,business ,Ex vivo ,medicine.drug - Abstract
OBJECTIVE Although oral methotrexate (MTX) remains the anchor drug for rheumatoid arthritis (RA), up to 50% of patients do not achieve a clinically adequate outcome. In addition, there is a lack of prognostic tools for treatment response prior to drug initiation. This study was undertaken to investigate whether interindividual differences in the human gut microbiome can aid in the prediction of MTX efficacy in new-onset RA. METHODS We performed 16S ribosomal RNA gene and shotgun metagenomic sequencing on the baseline gut microbiomes of drug-naive patients with new-onset RA (n = 26). Results were validated in an additional independent cohort (n = 21). To gain insight into potential microbial mechanisms, we conducted ex vivo experiments coupled with metabolomics analysis to evaluate the association between microbiome-driven MTX depletion and clinical response. RESULTS Our analysis revealed significant associations of the abundance of gut bacterial taxa and their genes with future clinical response (q < 0.05), including orthologs related to purine and MTX metabolism. Machine learning techniques were applied to the metagenomic data, resulting in a microbiome-based model that predicted lack of response to MTX in an independent group of patients. Finally, MTX levels remaining after ex vivo incubation with distal gut samples from pretreatment RA patients significantly correlated with the magnitude of future clinical response, suggesting a possible direct effect of the gut microbiome on MTX metabolism and treatment outcomes. CONCLUSION Taken together, these findings are the first step toward predicting lack of response to oral MTX in patients with new-onset RA and support the value of the gut microbiome as a possible prognostic tool and as a potential target in RA therapeutics.
- Published
- 2020
97. Differences in the Gut Microbiomes of Distinct Ethnicities Within the Same Geographic Area Are Linked to Host Metabolic Health
- Author
-
Eliseo Barajas, Suneil K. Koliwad, Ho Lim Lee, Andrew D. Patterson, Peter J. Turnbaugh, Jordan E. Bisanz, Grace Wei, Qi Yan Ang, Diana L. Alba, Cecilia Noecker, Vaibhav Upadhyay, and Jingwei Cai
- Subjects
biology ,Metagenomics ,Host (biology) ,Zoology ,Bacteroidetes ,Context (language use) ,Microbiome ,Species richness ,Gut flora ,biology.organism_classification ,Akkermansia muciniphila - Abstract
BackgroundThe human gut microbiota exhibits marked variation around the world, which has been attributed to dietary intake and other environmental factors. However, the degree to which ethnicity-associated differences in gut microbial community structure and function are maintained following immigration or in the context of metabolic disease is poorly understood.ResultsWe conducted a multi-omic study of 46 lean and obese East Asian and White participants living in the San Francisco Bay Area. 16S rRNA gene sequencing revealed significant differences between ethnic groups in bacterial richness and community structure. White individuals were enriched for the mucin-degrading Akkermansia muciniphila. East Asian participants had increased levels of multiple bacterial phyla, fermentative pathways detected by metagenomics, and the short-chain fatty acid end products acetate, propionate, and isobutyrate. Differences in the gut microbiota between the East Asian and White groups could not be explained by reported dietary intake, were more pronounced in lean individuals, and were associated with current geographical location. Microbiome transplantations into germ-free mice confirmed that the differences in the gut microbiota of the East Asian and White individuals we analyzed are independent of diet and that they differentially impact host body weight and adiposity in genetically identical mouse recipients.ConclusionsThe reported findings emphasize the utility of studying diverse ethnic groups within a defined geographical location and provide a starting point for dissecting the mechanisms contributing to the complex interactions between the gut microbiome and ethnicity-associated lifestyle, demographic, metabolic, and genetic factors.
- Published
- 2020
98. How gut microbiome interactions affect nutritional traits of Drosophila melanogaster
- Author
-
Angela E. Douglas, Jingwei Cai, Andrew D. Patterson, Grace Peters-Schulze, and John G. McMullen
- Subjects
Genetics ,0303 health sciences ,biology ,Physiology ,Context (language use) ,Aquatic Science ,medicine.disease_cause ,biology.organism_classification ,Yeast ,03 medical and health sciences ,0302 clinical medicine ,Microbial population biology ,Insect Science ,Acetobacter fabarum ,Lactobacillus ,medicine ,Animal Science and Zoology ,Fermentation ,Acetobacter ,Molecular Biology ,Drosophila ,030217 neurology & neurosurgery ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology - Abstract
Most research on the impact of the gut microbiome on animal nutrition is designed to identify the effects of single microbial taxa and single metabolites of microbial origin, without considering the potentially complex network of interactions among co-occurring microorganisms. Here, we investigated how different microbial associations and their fermentation products affect host nutrition, using Drosophila melanogaster colonized with three gut microorganisms (the bacteria Acetobacter fabarum and Lactobacillus brevis, and the yeast Hanseniaspora uvarum) in all seven possible combinations. Some microbial effects on host traits could be attributed to single taxa (e.g. yeast-mediated reduction of insect development time), while other effects were sex specific and driven by among-microbe interactions (e.g. male lipid content determined by interactions between the yeast and both bacteria). Parallel analysis of nutritional indices of microbe-free flies administered different microbial fermentation products (acetic acid, acetoin, ethanol and lactic acid) revealed a single consistent effect: that the lipid content of both male and female flies is reduced by acetic acid. This effect was recapitulated in male flies colonized with both yeast and A. fabarum, but not for any microbial treatment in females or males with other microbial complements. These data suggest that the effect of microbial fermentation products on host nutritional status is strongly context dependent, with respect to both the combination of associated microorganisms and host sex. Taken together, our findings demonstrate that among-microbe interactions can play a critically important role in determining the physiological outcome of host–microbiome interactions in Drosophila and, likely, in other animal hosts.
- Published
- 2020
99. Quantitative Analysis of Bile Acid with UHPLC-MS/MS
- Author
-
Philip B. Smith, Erik L. Allman, Yuan Tian, Andrew D. Patterson, and Jingwei Cai
- Subjects
0303 health sciences ,Cell signaling ,Chromatography ,Bile acid ,biology ,medicine.drug_class ,Chemistry ,010401 analytical chemistry ,Gut flora ,Tandem mass spectrometry ,biology.organism_classification ,digestive system ,01 natural sciences ,Uhplc ms ms ,0104 chemical sciences ,03 medical and health sciences ,Metabolomics ,medicine ,Cholesterol metabolism ,Quantitative analysis (chemistry) ,030304 developmental biology - Abstract
Bile acids are important end products of cholesterol metabolism, having been shown to serve as signaling molecules and intermediates between the host and the gut microbiota. Here we describe a robust and accurate method using ultrahigh-pressure liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS) for the quantification of bile acids in stool/cecal and tissue samples.
- Published
- 2020
100. Sample Preparation and Data Analysis for NMR-Based Metabolomics
- Author
-
Andrew D. Patterson, Yuan Tian, and Tapas K. Mal
- Subjects
0303 health sciences ,Chromatography ,Metabolite ,010401 analytical chemistry ,Nuclear magnetic resonance spectroscopy ,01 natural sciences ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Metabolomics ,Heteronuclear molecule ,chemistry ,Proton NMR ,Sample preparation ,Biomarker discovery ,Heteronuclear single quantum coherence spectroscopy ,030304 developmental biology - Abstract
NMR spectroscopy has become one of the preferred analytical techniques for metabolomics studies due to its inherent nondestructive nature, ability to identify and quantify metabolites simultaneously in a complex mixture, minimal sample preparation requirement, and high degree of experimental reproducibility. NMR-based metabolomics studies involve the measurement and multivariate statistical analysis of metabolites present in biological samples such as biofluids, stool/feces, intestinal content, tissue, and cell extracts by high-resolution NMR spectroscopy-the goal then is to identify and quantify metabolites and evaluate changes of metabolite concentrations in response to some perturbation. Here we describe methodologies for NMR sample preparation of biofluids (serum, saliva, and urine) and stool/feces, intestinal content, and tissues for NMR experiments including extraction of polar metabolites and application of NMR in metabolomics studies. One dimensional (1D) 1H NMR experiments with different variations such as pre-saturation, relaxation-edited, and diffusion-edited are routinely acquired for profiling and metabolite identification and quantification. 2D homonuclear 1H-1H TOCSY and COSY, 2D J-resolved, and heteronuclear 1H-13C HSQC and HMBC are also performed to assist with metabolite identification and quantification. The NMR data are then subjected to targeted and/or untargeted multivariate statistical analysis for biomarker discovery, clinical diagnosis, toxicological studies, molecular phenotyping, and functional genomics.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.