Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Seth D. Rhoades is active.

Publication


Featured researches published by Seth D. Rhoades.


Journal of Biological Chemistry | 2015

Independent Effects of γ-Aminobutyric Acid Transaminase (GABAT) on Metabolic and Sleep Homeostasis.

Sarah E. Maguire; Seth D. Rhoades; Wen-Feng Chen; Arjun Sengupta; Zhifeng Yue; Jason C. Lim; Claire H. Mitchell; Aalim M. Weljie; Amita Sehgal

Background: Components of GABA catabolism feed into sleep and potential energy pathways. Results: We identified a metabolic phenotype in Drosophila mutants of GABA turnover and traced it to a limit in glutamate, which is not relevant for sleep. Conclusion: GABA regulates metabolic and sleep homeostasis through independent mechanisms. Significance: Neurological disorders involving GABA disruption may be associated with metabolic problems. Breakdown of the major sleep-promoting neurotransmitter, γ-aminobutyric acid (GABA), in the GABA shunt generates catabolites that may enter the tricarboxylic acid cycle, but it is unknown whether catabolic by-products of the GABA shunt actually support metabolic homeostasis. In Drosophila, the loss of the specific enzyme that degrades GABA, GABA transaminase (GABAT), increases sleep, and we show here that it also affects metabolism such that flies lacking GABAT fail to survive on carbohydrate media. Expression of GABAT in neurons or glia rescues this phenotype, indicating a general metabolic function for this enzyme in the brain. As GABA degradation produces two catabolic products, glutamate and succinic semialdehyde, we sought to determine which was responsible for the metabolic phenotype. Through genetic and pharmacological experiments, we determined that glutamate, rather than succinic semialdehyde, accounts for the metabolic phenotype of gabat mutants. This is supported by biochemical measurements of catabolites in wild-type and mutant animals. Using in vitro labeling assays, we found that inhibition of GABAT affects energetic pathways. Interestingly, we also observed that gaba mutants display a general disruption in bioenergetics as measured by altered levels of tricarboxylic acid cycle intermediates, NAD+/NADH, and ATP levels. Finally, we report that the effects of GABAT on sleep do not depend upon glutamate, indicating that GABAT regulates metabolic and sleep homeostasis through independent mechanisms. These data indicate a role of the GABA shunt in the development of metabolic risk and suggest that neurological disorders caused by altered glutamate or GABA may be associated with metabolic disruption.


Scientific Reports | 2017

A Pilot Characterization of the Human Chronobiome

Carsten Skarke; Nicholas F. Lahens; Seth D. Rhoades; Amy E. Campbell; Kyle Bittinger; Aubrey Bailey; Christian Hoffmann; Randal S. Olson; Lihong Chen; Guangrui Yang; Thomas S. Price; Jason H. Moore; Frederic D. Bushman; Casey S. Greene; Gregory R. Grant; Aalim M. Weljie; Garret A. FitzGerald

Physiological function, disease expression and drug effects vary by time-of-day. Clock disruption in mice results in cardio-metabolic, immunological and neurological dysfunction; circadian misalignment using forced desynchrony increases cardiovascular risk factors in humans. Here we integrated data from remote sensors, physiological and multi-omics analyses to assess the feasibility of detecting time dependent signals - the chronobiome – despite the “noise” attributable to the behavioral differences of free-living human volunteers. The majority (62%) of sensor readouts showed time-specific variability including the expected variation in blood pressure, heart rate, and cortisol. While variance in the multi-omics is dominated by inter-individual differences, temporal patterns are evident in the metabolome (5.4% in plasma, 5.6% in saliva) and in several genera of the oral microbiome. This demonstrates, despite a small sample size and limited sampling, the feasibility of characterizing at scale the human chronobiome “in the wild”. Such reference data at scale are a prerequisite to detect and mechanistically interpret discordant data derived from patients with temporal patterns of disease expression, to develop time-specific therapeutic strategies and to refine existing treatments.


The International Journal of Biochemistry & Cell Biology | 2017

Sleep restriction induced energy, methylation and lipogenesis metabolic switches in rat liver

Arjun Sengupta; Seth D. Rhoades; Eun Ji Kim; Soumyashant Nayak; Gregory R. Grant; Peter Meerlo; Aalim M. Weljie

Sleep curtailment is ubiquitous in modern day society. Sleep debt is associated with maladaptive physiological changes that can lead to cardiometabolic and neuropsychiatric pathologies. Recent literature has shown the effects of sleep restriction (SR) on systemic metabolic profiles in biofluids, implying that tissue-specific metabolomes are impacted by SR. To test this hypothesis, we assessed hepatic metabolic profiles of rats after 5days of SR using UPLC-MS based metabolomics analysis and gene expression analysis. Our data suggests distinctive effects of SR on the liver metabolic profile of rats compared to forced-activity control animals. We observed specific impacts of SR on NAD metabolism through NAD accumulation and upregulation of Nampt, the rate determining step of NAD salvage. Additional multi-omic changes were observed in methionine metabolism, with an elevated SAM:SAH ratio under SR. This effect on one carbon metabolism is indicative of increased methylation potential. Changes in TCA cycle intermediates and ATP-citrate lyase (Acly) gene expression were observed that may be related to altered circulatory lipid profiles previously reported documenting the chrono-metabolic connection. Taken together with previous investigations, these observations are consistent with a model of decreased TCA activity with concomitant increase in lipogenesis induced by SR. These tissue-specific mechanistic insights into metabolic effects of SR provide a springboard to future metabolic intervention studies.


Current Opinion in Biotechnology | 2017

Time is ripe: maturation of metabolomics in chronobiology.

Seth D. Rhoades; Arjun Sengupta; Aalim M. Weljie

Sleep and circadian rhythms studies have recently benefited from metabolomics analyses, uncovering new connections between chronobiology and metabolism. From untargeted mass spectrometry to quantitative nuclear magnetic resonance spectroscopy, a diversity of analytical approaches has been applied for biomarker discovery in the field. In this review we consider advances in the application of metabolomics technologies which have uncovered significant effects of sleep and circadian cycles on several metabolites, namely phosphatidylcholine species, medium-chain carnitines, and aromatic amino acids. Study design and data processing measures essential for detecting rhythmicity in metabolomics data are also discussed. Future developments in these technologies are anticipated vis-à-vis validating early findings, given metabolomics has only recently entered the ring with other systems biology assessments in chronometabolism studies.


Journal of Biological Rhythms | 2018

Circadian- and Light-driven Metabolic Rhythms in Drosophila melanogaster:

Seth D. Rhoades; Katrina Nayak; Shirley L. Zhang; Amita Sehgal; Aalim M. Weljie

Complex interactions of environmental cues and transcriptional clocks drive rhythmicity in organismal physiology. Light directly affects the circadian clock; however, little is known about its relative role in controlling metabolic variations in vivo. Here we used high time-resolution sampling in Drosophila at every 2 h to measure metabolite outputs using a liquid-chromatography tandem mass spectrometry (LC-MS/MS) approach. Over 14% of detected metabolites oscillated with circadian periodicity under light-dark (LD) cycles. Many metabolites peaked shortly after lights-on, suggesting responsiveness to feeding and/or activity rather than the preactivity anticipation, as observed in previous transcriptomics analyses. Roughly 9% of measured metabolites uniquely oscillated under constant darkness (DD), suggesting that metabolite rhythms are associated with the transcriptional clock machinery. Strikingly, metabolome differences between LD and constant darkness were observed only during the light phase, highlighting the importance of photic input. Clock mutant flies exhibited strong 12-h ultradian rhythms, including 4 carbohydrate species with circadian periods in wild-type flies, but lacked 24-h circadian metabolic oscillations. A meta-analysis of these results with previous circadian metabolomics experiments uncovered the possibility of conserved rhythms in amino acids, keto-acids, and sugars across flies, mice, and humans and provides a basis for exploring the chrono-metabolic connection with powerful genetic tools in Drosophila.


Circulation | 2018

Cyclooxygenase-2, Asymmetric Dimethylarginine and the Cardiovascular Hazard from NSAIDs

Emanuela Ricciotti; Cecilia Castro; Soon Yew Tang; William T. E. Briggs; James A. West; Dania Malik; Seth D. Rhoades; Hu Meng; Xuanwen Li; Nicholas F. Lahens; Jeffrey A. Sparks; Elizabeth W. Karlson; Aalim M. Weljie; Julian L. Griffin; Garret A. FitzGerald

Background: Large-scale, placebo-controlled trials established that nonsteroidal anti-inflammatory drugs confer a cardiovascular hazard: this has been attributed to depression of cardioprotective products of cyclooxygenase (COX)–2, especially prostacyclin. An alternative mechanism by which nonsteroidal anti-inflammatory drugs might constrain cardioprotection is by enhancing the formation of methylarginines in the kidney that would limit the action of nitric oxide throughout the vasculature. Methods: Targeted and untargeted metabolomics were used to investigate the effect of COX-2 deletion or inhibition in mice and in osteoarthritis patients exposed to nonsteroidal anti-inflammatory drugs on the L-arginine/nitric oxide pathway. Results: Analysis of the plasma and renal metabolome was performed in postnatal tamoxifen-inducible Cox-2 knockout mice, which exhibit normal renal function and blood pressure. This revealed no changes in arginine and methylarginines compared with their wild-type controls. Moreover, the expression of genes in the L-arginine/nitric oxide pathway was not altered in the renal medulla or cortex of tamoxifen inducible Cox-2 knockout mice. Therapeutic concentrations of the selective COX-2 inhibitors, rofecoxib, celecoxib, and parecoxib, none of which altered basal blood pressure or renal function as reflected by plasma creatinine, failed to elevate plasma arginine and methylarginines in mice. Finally, plasma arginine or methylarginines were not altered in osteoarthritis patients with confirmed exposure to nonsteroidal anti-inflammatory drugs that inhibit COX-1 and COX-2. By contrast, plasma asymmetrical dimethylarginine was increased in mice infused with angiotensin II sufficient to elevate blood pressure and impair renal function. Four weeks later, blood pressure, plasma creatinine, and asymmetrical dimethylarginine were restored to normal levels. The increase in asymmetrical dimethylarginine in response to infusion with angiotensin II in celecoxib-treated mice was also related to transient impairment of renal function. Conclusions: Plasma methylarginines are not altered by COX-2 deletion or inhibition but rather are elevated coincident with renal compromise.


Cell Metabolism | 2017

Clock Regulation of Metabolites Reveals Coupling between Transcription and Metabolism

Saikumari Y. Krishnaiah; Gang Wu; Brian J. Altman; Jacqueline Growe; Seth D. Rhoades; Faith Coldren; Anand Venkataraman; Anthony O. Olarerin-George; Lauren J. Francey; Sarmistha Mukherjee; Saiveda Girish; Christopher P. Selby; Sibel Cal; Ubeydullah Er; Bahareh Sianati; Arjun Sengupta; Ron C. Anafi; I. Halil Kavakli; Aziz Sancar; Joseph A. Baur; Chi V. Dang; John B. Hogenesch; Aalim M. Weljie


Molecular Cell | 2017

Nuclear Acetyl-CoA Production by ACLY Promotes Homologous Recombination

Sharanya Sivanand; Seth D. Rhoades; Qinqin Jiang; Joyce V. Lee; Joseph L. Benci; Jingwen Zhang; Salina Yuan; Isabella Viney; Steven Zhao; Alessandro Carrer; Michael Bennett; Andy J. Minn; Aalim M. Weljie; Roger A. Greenberg; Kathryn E. Wellen


Metabolomics | 2016

Comprehensive optimization of LC–MS metabolomics methods using design of experiments (COLMeD)

Seth D. Rhoades; Aalim M. Weljie


Cell Metabolism | 2017

Erratum: Clock Regulation of Metabolites Reveals Coupling between Transcription and Metabolism (Cell Metabolism (2017) 25(4) (961–974.e4) (S1550413117301717) (10.1016/j.cmet.2017.03.019))

Saikumari Y. Krishnaiah; Gang Wu; Brian J. Altman; Jacqueline Growe; Seth D. Rhoades; Faith Coldren; Anand Venkataraman; Anthony O. Olarerin-George; Lauren J. Francey; Sarmistha Mukherjee; Saiveda Girish; Christopher P. Selby; Sibel Cal; Ubeydullah Er; Bahareh Sianati; Arjun Sengupta; Ron C. Anafi; I. Halil Kavakli; Aziz Sancar; Joseph A. Baur; Chi V. Dang; John B. Hogenesch; Aalim M. Weljie

Collaboration


Dive into the Seth D. Rhoades's collaboration.

Top Co-Authors

Avatar

Aalim M. Weljie

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Arjun Sengupta

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Amita Sehgal

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aziz Sancar

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Bahareh Sianati

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brian J. Altman

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Chi V. Dang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christopher P. Selby

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge