29 results on '"Willis, Craig R. G."'
Search Results
2. How to obtain an integrated picture of the molecular networks involved in adaptation to microgravity in different biological systems?
- Author
-
Willis, Craig R. G., Calvaruso, Marco, Angeloni, Debora, Baatout, Sarah, Benchoua, Alexandra, Bereiter-Hahn, Juergen, Bottai, Daniele, Buchheim, Judith-Irina, Carnero-Diaz, Eugénie, Castiglioni, Sara, Cavalieri, Duccio, Ceccarelli, Gabriele, Chouker, Alexander, Cialdai, Francesca, Ciofani, Gianni, Coppola, Giuseppe, Cusella, Gabriella, Degl’Innocenti, Andrea, Desaphy, Jean-Francois, Frippiat, Jean-Pol, Gelinsky, Michael, Genchi, Giada, Grano, Maria, Grimm, Daniela, Guignandon, Alain, Herranz, Raúl, Hellweg, Christine, Iorio, Carlo Saverio, Karapantsios, Thodoris, van Loon, Jack, Lulli, Matteo, Maier, Jeanette, Malda, Jos, Mamaca, Emina, Morbidelli, Lucia, Osterman, Andreas, Ovsianikov, Aleksandr, Pampaloni, Francesco, Pavezlorie, Elizabeth, Pereda-Campos, Veronica, Przybyla, Cyrille, Rettberg, Petra, Rizzo, Angela Maria, Robson-Brown, Kate, Rossi, Leonardo, Russo, Giorgio, Salvetti, Alessandra, Risaliti, Chiara, Santucci, Daniela, Sperl, Matthias, Tabury, Kevin, Tavella, Sara, Thielemann, Christiane, Willaert, Ronnie, Monici, Monica, and Szewczyk, Nathaniel J.
- Published
- 2024
- Full Text
- View/download PDF
3. Proteomic features of skeletal muscle adaptation to resistance exercise training as a function of age
- Author
-
Deane, Colleen S., Phillips, Bethan E., Willis, Craig R. G., Wilkinson, Daniel J., Smith, Ken, Higashitani, Nahoko, Williams, John P., Szewczyk, Nathaniel J., Atherton, Philip J., Higashitani, Atsushi, and Etheridge, Timothy
- Published
- 2023
- Full Text
- View/download PDF
4. Myokines, Measurement, and Technical Considerations
- Author
-
Willis, Craig R. G., primary, Deane, Colleen S., additional, and Etheridge, Timothy, additional
- Published
- 2023
- Full Text
- View/download PDF
5. How to obtain an integrated picture of the molecular networks involved in adaptation to microgravity in different biological systems?
- Author
-
Willis, Craig R G, Calvaruso, Marco, Angeloni, Debora, Baatout, Sarah, Benchoua, Alexandra, Bereiter-Hahn, Juergen, Bottai, Daniele, Buchheim, Judith-Irina, Carnero-Diaz, Eugénie, Castiglioni, Sara, Cavalieri, Duccio, Ceccarelli, Gabriele, Chouker, Alexander, Cialdai, Francesca, Ciofani, Gianni, Coppola, Giuseppe, Cusella, Gabriella, Degl'Innocenti, Andrea, Desaphy, Jean-Francois, Frippiat, Jean-Pol, Gelinsky, Michael, Genchi, Giada, Grano, Maria, Grimm, Daniela, Guignandon, Alain, Herranz, Raúl, Hellweg, Christine, Iorio, Carlo Saverio, Karapantsios, Thodoris, van Loon, Jack, Lulli, Matteo, Maier, Jeanette, Malda, Jos, Mamaca, Emina, Morbidelli, Lucia, Osterman, Andreas, Ovsianikov, Aleksandr, Pampaloni, Francesco, Pavezlorie, Elizabeth, Pereda-Campos, Veronica, Przybyla, Cyrille, Rettberg, Petra, Rizzo, Angela Maria, Robson-Brown, Kate, Rossi, Leonardo, Russo, Giorgio, Salvetti, Alessandra, Risaliti, Chiara, Santucci, Daniela, Sperl, Matthias, Tabury, Kevin, Tavella, Sara, Thielemann, Christiane, Willaert, Ronnie, Monici, Monica, Szewczyk, Nathaniel J, Willis, Craig R G, Calvaruso, Marco, Angeloni, Debora, Baatout, Sarah, Benchoua, Alexandra, Bereiter-Hahn, Juergen, Bottai, Daniele, Buchheim, Judith-Irina, Carnero-Diaz, Eugénie, Castiglioni, Sara, Cavalieri, Duccio, Ceccarelli, Gabriele, Chouker, Alexander, Cialdai, Francesca, Ciofani, Gianni, Coppola, Giuseppe, Cusella, Gabriella, Degl'Innocenti, Andrea, Desaphy, Jean-Francois, Frippiat, Jean-Pol, Gelinsky, Michael, Genchi, Giada, Grano, Maria, Grimm, Daniela, Guignandon, Alain, Herranz, Raúl, Hellweg, Christine, Iorio, Carlo Saverio, Karapantsios, Thodoris, van Loon, Jack, Lulli, Matteo, Maier, Jeanette, Malda, Jos, Mamaca, Emina, Morbidelli, Lucia, Osterman, Andreas, Ovsianikov, Aleksandr, Pampaloni, Francesco, Pavezlorie, Elizabeth, Pereda-Campos, Veronica, Przybyla, Cyrille, Rettberg, Petra, Rizzo, Angela Maria, Robson-Brown, Kate, Rossi, Leonardo, Russo, Giorgio, Salvetti, Alessandra, Risaliti, Chiara, Santucci, Daniela, Sperl, Matthias, Tabury, Kevin, Tavella, Sara, Thielemann, Christiane, Willaert, Ronnie, Monici, Monica, and Szewczyk, Nathaniel J
- Abstract
Periodically, the European Space Agency (ESA) updates scientific roadmaps in consultation with the scientific community. The ESA SciSpacE Science Community White Paper (SSCWP) 9, "Biology in Space and Analogue Environments", focusses in 5 main topic areas, aiming to address key community-identified knowledge gaps in Space Biology. Here we present one of the identified topic areas, which is also an unanswered question of life science research in Space: "How to Obtain an Integrated Picture of the Molecular Networks Involved in Adaptation to Microgravity in Different Biological Systems?" The manuscript reports the main gaps of knowledge which have been identified by the community in the above topic area as well as the approach the community indicates to address the gaps not yet bridged. Moreover, the relevance that these research activities might have for the space exploration programs and also for application in industrial and technological fields on Earth is briefly discussed.
- Published
- 2024
6. Transcriptomic adaptation during skeletal muscle habituation to eccentric or concentric exercise training
- Author
-
Willis, Craig R. G., Deane, Colleen S., Ames, Ryan M., Bass, Joseph J., Wilkinson, Daniel J., Smith, Kenneth, Phillips, Bethan E., Szewczyk, Nathaniel J., Atherton, Philip J., and Etheridge, Timothy
- Published
- 2021
- Full Text
- View/download PDF
7. Spaceflight Induces Strength Decline in Caenorhabditis elegans
- Author
-
Soni, Purushottam, primary, Edwards, Hunter, additional, Anupom, Taslim, additional, Rahman, Mizanur, additional, Lesanpezeshki, Leila, additional, Blawzdziewicz, Jerzy, additional, Cope, Henry, additional, Gharahdaghi, Nima, additional, Scott, Daniel, additional, Toh, Li Shean, additional, Williams, Philip M., additional, Etheridge, Timothy, additional, Szewczyk, Nathaniel, additional, Willis, Craig R. G., additional, and Vanapalli, Siva A., additional
- Published
- 2023
- Full Text
- View/download PDF
8. Mitochondrial sulfide promotes life span and health span through distinct mechanisms in developing versus adult treated Caenorhabditis elegans
- Author
-
Vintila, Adriana Raluca, primary, Slade, Luke, additional, Cooke, Michael, additional, Willis, Craig R. G., additional, Torregrossa, Roberta, additional, Rahman, Mizanur, additional, Anupom, Taslim, additional, Vanapalli, Siva A., additional, Gaffney, Christopher J., additional, Gharahdaghi, Nima, additional, Szabo, Csaba, additional, Szewczyk, Nathaniel J., additional, Whiteman, Matthew, additional, and Etheridge, Timothy, additional
- Published
- 2023
- Full Text
- View/download PDF
9. Mitochondrial sulfide promotes life span and health span through distinct mechanisms in developing versus adult treated Caenorhabditis elegans
- Author
-
Vintila, Adriana Raluca, Slade, Luke, Cooke, Michael, Willis, Craig R. G., Torregrossa, Roberta, Rahman, Mizanur, Anupom, Taslim, Vanapalli, Siva A., Gaffney, Christopher J., Gharahdaghi, Nima, Szabo, Csaba, Szewczyk, Nathaniel J., Whiteman, Matthew, Etheridge, Timothy, Vintila, Adriana Raluca, Slade, Luke, Cooke, Michael, Willis, Craig R. G., Torregrossa, Roberta, Rahman, Mizanur, Anupom, Taslim, Vanapalli, Siva A., Gaffney, Christopher J., Gharahdaghi, Nima, Szabo, Csaba, Szewczyk, Nathaniel J., Whiteman, Matthew, and Etheridge, Timothy
- Abstract
Living longer without simultaneously extending years spent in good health ("health span") is an increasing societal burden, demanding new therapeutic strategies. Hydrogen sulfide (H S) can correct disease-related mitochondrial metabolic deficiencies, and supraphysiological H S concentrations can pro health span. However, the efficacy and mechanisms of mitochondrion-targeted sulfide delivery molecules (mtH S) administered across the adult life course are unknown. Using a aging model, we compared untargeted H S (NaGYY4137, 100 µM and 100 nM) and mtH S (AP39, 100 nM) donor effects on life span, neuromuscular health span, and mitochondrial integrity. H S donors were administered from birth or in young/middle-aged animals (day 0, 2, or 4 postadulthood). RNAi pharmacogenetic interventions and transcriptomics/network analysis explored molecular events governing mtH S donor-mediated health span. Developmentally administered mtH S (100 nM) improved life/health span vs. equivalent untargeted H S doses. mtH S preserved aging mitochondrial structure, content (citrate synthase activity) and neuromuscular strength. Knockdown of H S metabolism enzymes and FoxO/ prevented the positive health span effects of mtH S, whereas DCAF11/ - Nrf2/ oxidative stress protection pathways were dispensable. Health span, but not life span, increased with all adult-onset mtH S treatments. Adult mtH S treatment also rejuvenated aging transcriptomes by minimizing expression declines of mitochondria and cytoskeletal components, and peroxisome metabolism hub components, under mechanistic control by the / transcription factor circuit. H S health span extension likely acts at the mitochondrial level, the mechanisms of which dissociate from life span across adult vs. developmental treatment timings. The small mtH S doses required for health span extension, combined with efficacy in adult animals, suggest mtH S is a potential healthy aging therapeutic.
- Published
- 2023
10. RNA-Sequencing Muscle Plasticity to Resistance Exercise Training and Disuse in Youth and Older Age
- Author
-
Fernandez-Gonzalo, Rodrigo, primary, Willis, Craig R. G., additional, Etheridge, Timothy, additional, and Deane, Colleen S., additional
- Published
- 2022
- Full Text
- View/download PDF
11. Proteomic features of skeletal muscle adaptation to resistance exercise training as a function of age
- Author
-
Deane, Colleen S., primary, Phillips, Bethan E., additional, Willis, Craig R. G., additional, Wilkinson, Daniel J., additional, Smith, Ken, additional, Higashitani, Nahoko, additional, Williams, John P., additional, Szewczyk, Nathaniel J., additional, Atherton, Philip J., additional, Higashitani, Atsushi, additional, and Etheridge, Timothy, additional
- Published
- 2022
- Full Text
- View/download PDF
12. Promotion of Joint Degeneration and Chondrocyte Metabolic Dysfunction by Excessive Growth Hormone in Mice.
- Author
-
Zhu, Shouan, Liu, Huanhuan, Davis, Trent, Willis, Craig R. G., Basu, Reetobrata, Witzigreuter, Luke, Bell, Stephen, Szewczyk, Nathaniel, Lotz, Martin K., Hill, Marcheta, Fajardo, Roberto J., O'Connor, Patrick M., Berryman, Darlene E., and Kopchick, John J.
- Subjects
ALANINE metabolism ,TRYPTOPHAN metabolism ,LYSINE metabolism ,KNEE joint ,CARTILAGE cells ,KNEE osteoarthritis ,SYNOVITIS ,ANIMAL experimentation ,METABOLOMICS ,RISK assessment ,CYTOCHEMISTRY ,GENE expression ,RESEARCH funding ,ARTICULAR cartilage ,PITUITARY hormones ,COMPUTED tomography ,OXIDOREDUCTASES ,MICE ,OXIDATION-reduction reaction ,FATTY acids ,DISEASE risk factors - Abstract
Objective: Many patients with acromegaly, a hormonal disorder with excessive growth hormone (GH) production, report pain in joints. We undertook this study to characterize the joint pathology of mice with overexpression of bovine GH (bGH) or a GH receptor antagonist (GHa) and to investigate the effect of GH on regulation of chondrocyte cellular metabolism. Methods: Knee joints from mice overexpressing bGH or GHa and wild‐type (WT) control mice were examined using histology and micro–computed tomography for osteoarthritic (OA) pathologies. Additionally, cartilage from bGH mice was used for metabolomics analysis. Mouse primary chondrocytes from bGH and WT mice, with or without pegvisomant treatment, were used for quantitative polymerase chain reaction and Seahorse respirometry analyses. Results: Both male and female bGH mice at ~13 months of age had increased knee joint degeneration, which was characterized by loss of cartilage structure, expansion of hypertrophic chondrocytes, synovitis, and subchondral plate thinning. The joint pathologies were also demonstrated by significantly higher Osteoarthritis Research Society International and Mankin scores in bGH mice compared to WT control mice. Metabolomics analysis revealed changes in a wide range of metabolic pathways in bGH mice, including beta‐alanine metabolism, tryptophan metabolism, lysine degradation, and ascorbate and aldarate metabolism. Also, bGH chondrocytes up‐regulated fatty acid oxidation and increased expression of Col10a. Joints of GHa mice were remarkably protected from developing age‐associated joint degeneration, with smooth articular joint surface. Conclusion: This study showed that an excessive amount of GH promotes joint degeneration in mice, which was associated with chondrocyte metabolic dysfunction and hypertrophic changes, whereas antagonizing GH action through a GHa protects mice from OA development. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Routine omics collection is a golden opportunity for European human research in space and analog environments
- Author
-
Cope, Henry, Willis, Craig R. G., MacKay, Matthew J., Rutter, Lindsay A., Toh, Li Shean, Williams, Philip M., Herranz, Raul, Borg, Joseph, Bezdan, Daniela, Giacomello, Stefania, Muratani, Masafumi, Mason, Christopher E., Etheridge, Timothy, Szewczyk, Nathaniel J., Cope, Henry, Willis, Craig R. G., MacKay, Matthew J., Rutter, Lindsay A., Toh, Li Shean, Williams, Philip M., Herranz, Raul, Borg, Joseph, Bezdan, Daniela, Giacomello, Stefania, Muratani, Masafumi, Mason, Christopher E., Etheridge, Timothy, and Szewczyk, Nathaniel J.
- Abstract
Widespread generation and analysis of omics data have revolutionized molecular medicine on Earth, yet its power to yield new mechanistic insights and improve occupational health during spaceflight is still to be fully realized in humans. Nevertheless, rapid technological advancements and ever-regular spaceflight programs mean that longitudinal, standardized, and cost-effective collection of human space omics data are firmly within reach. Here, we consider the practicality and scientific return of different sampling methods and omic types in the context of human spaceflight, We also appraise ethical and legal considerations pertinent to omics data derived from European astronauts and spaceflight participants (SFPs). Ultimately, we propose that a routine omics collection program in spaceflight and analog environments presents a golden opportunity. Unlocking this bright future of artificial intelligence (AI)-driven analyses and personalized medicine approaches will require further investigation into best practices, including policy design and standardization of omics data, metadata, and sampling methods., QC 20230113
- Published
- 2022
- Full Text
- View/download PDF
14. Transcriptomic links to muscle mass loss and declines in cumulative muscle protein synthesis during short‐term disuse in healthy younger humans
- Author
-
Willis, Craig R. G., primary, Gallagher, Iain J., additional, Wilkinson, Daniel J., additional, Brook, Matthew S., additional, Bass, Joseph J., additional, Phillips, Bethan E., additional, Smith, Kenneth, additional, Etheridge, Timothy, additional, Stokes, Tanner, additional, McGlory, Chris, additional, Gorissen, Stefan H. M., additional, Szewczyk, Nathaniel J., additional, Phillips, Stuart M., additional, and Atherton, Philip J., additional
- Published
- 2021
- Full Text
- View/download PDF
15. Nrf2 deficiency induces skeletal muscle mitochondrial dysfunction: a proteomics/bioinformatics approach
- Author
-
Willis, Craig R. G., primary and Deane, Colleen S., additional
- Published
- 2020
- Full Text
- View/download PDF
16. Comparative Transcriptomics Identifies Altered Neuronal and Metabolic Function as Common Adaptations to Microgravity and Hypergravity in Caenorhabditis elegans
- Author
-
Willis, Craig R. G., primary, Szewczyk, Nathaniel J., additional, Costes, Sylvain V., additional, Udranszky, Ingrid A., additional, Reinsch, Sigrid S., additional, Etheridge, Timothy, additional, and Conley, Catharine A., additional
- Published
- 2020
- Full Text
- View/download PDF
17. Single cell transcriptomics identifies master regulators of dysfunctional pathways in SOD1 ALS motor neurons
- Author
-
Namboori, Seema C., Thomas, Patricia, Ames, Ryan, Garrett, Lawrence O., Willis, Craig R. G., Stanton, Lawrence W., and Bhinge, Akshay
- Subjects
nervous system - Abstract
Background Bulk RNA-Seq has been extensively utilized to investigate the molecular changes accompanying motor neuron degeneration in Amyotrophic Lateral Sclerosis (ALS). However, due to the heterogeneity and degenerating phenotype of the neurons, it has proved difficult to assign specific changes to neuronal subtypes and identify which factors drive these changes. Consequently, we have utilized single cell transcriptomics of degenerating motor neurons derived from ALS patients to uncover key transcriptional drivers of dysfunctional pathways. Results Single cell analysis of spinal neuronal cultures derived from ALS and isogenic iPSC allowed us to classify cells into neural subtypes including motor neurons and interneurons. Differential expression analysis between disease and control motor neurons revealed downregulation of genes involved in synaptic structure, neuromuscular junction, neuronal cytoskeleton and mitochondrial function. Interestingly, interneurons did not show similar suppression of these homeostatic functions. Single cell expression data enabled us to derive a context-specific transcriptional network relevant to ALS neurons. Master regulator analysis on this network identified core transcriptional factors driving the ALS disease signature. Specifically, we were able to correlate suppression of HOXA1 and HOXA5 to synaptic dysfunction in ALS motor neurons. Our results suggest that suppression of HOX genes may be a general phenomenon in SOD1 ALS. Conclusions Our results demonstrate the utility of single cell transcriptomics in mapping disease-relevant gene regulatory networks driving neurodegeneration in ALS motor neurons. We propose that ALS-associated mutant SOD1 leads to inhibition of transcriptional networks driving homeostatic programs specific to motor neurons, thereby providing a possible explanation for the relative resistance of spinal interneurons to degeneration in ALS.
- Published
- 2019
- Full Text
- View/download PDF
18. The acute transcriptional response to resistance exercise : impact of age and contraction mode
- Author
-
Deane, Colleen S., Ames, Ryan M., Phillips, Bethan E., Weedon, Michael N., Willis, Craig R. G., Boereboom, Catherine, Abdulla, Haitham, Bukhari, Syed S. I., Lund, Jonathan N., Williams, John P., Wilkinson, Daniel J., Smith, Kenneth, Gallagher, Iain J., Kadi, Fawzi, Szewczyk, Nathaniel J., Atherton, Philip J., Etheridge, Timothy, Deane, Colleen S., Ames, Ryan M., Phillips, Bethan E., Weedon, Michael N., Willis, Craig R. G., Boereboom, Catherine, Abdulla, Haitham, Bukhari, Syed S. I., Lund, Jonathan N., Williams, John P., Wilkinson, Daniel J., Smith, Kenneth, Gallagher, Iain J., Kadi, Fawzi, Szewczyk, Nathaniel J., Atherton, Philip J., and Etheridge, Timothy
- Abstract
Optimization of resistance exercise (RE) remains a hotbed of research for muscle building and maintenance. However, the interactions between the contractile components of RE (i.e. concentric (CON) and eccentric (ECC)) and age, are poorly defined. We used transcriptomics to compare age-related molecular responses to acute CON and ECC exercise. Eight young (21 +/- 1 y) and eight older (70 +/- 1 y) exercise-naive male volunteers had vastus lateralis biopsies collected at baseline and 5 h post unilateral CON and contralateral ECC exercise. RNA was subjected to next-generation sequencing and differentially expressed (DE) genes tested for pathway enrichment using Gene Ontology (GO). The young transcriptional response to CON and ECC was highly similar and older adults displayed moderate contraction-specific profiles, with no GO enrichment. Age-specific responses to ECC revealed 104 DE genes unique to young, and 170 DE genes in older muscle, with no GO enrichment. Following CON, 15 DE genes were young muscle-specific, whereas older muscle uniquely expressed 147 up-regulated genes enriched for cell adhesion and blood vessel development, and 28 down-regulated genes involved in mitochondria! respiration, amino acid and lipid metabolism. Thus, older age is associated with contraction-specific regulation often without clear functional relevance, perhaps reflecting a degree of stochastic age-related dysregulation., Funding Agencies:Bournemouth UniversityEPSRC/BBSRC Innovation Fellowship EP/S001352/1 Medical Research Council MR/P021220/1 MR/K00414X/1 Arthritis Research UK 19891 National Institute for Health Research, Nottingham Biomedical Research Centre Biotechnology and Biological Sciences Research Council BB/N015894/1 BB/J014400/1 BB/M009122/1 Swedish Research Council for Sport Science 2016/125 2017/143
- Published
- 2019
- Full Text
- View/download PDF
19. Single cell transcriptomics identifies master regulators of neurodegeneration in SOD1 ALS motor neurons
- Author
-
Namboori, Seema C., primary, Thomas, Patricia, additional, Ames, Ryan, additional, Hawkins, Sophie, additional, Garrett, Lawrence O., additional, Willis, Craig R. G., additional, Rosa, Alessandro, additional, Stanton, Lawrence W., additional, and Bhinge, Akshay, additional
- Published
- 2019
- Full Text
- View/download PDF
20. Nrf2 deficiency induces skeletal muscle mitochondrial dysfunction: a proteomics/bioinformatics approach.
- Author
-
Willis, Craig R. G. and Deane, Colleen S.
- Subjects
- *
SKELETAL muscle , *EPICATECHIN , *PROTEOMICS , *MITOCHONDRIA , *LIFE sciences , *MUSCLE fatigue , *ISOFLURANE - Abstract
Keywords: bioinformatics; mitochondria; proteomics; redox; skeletal muscle EN bioinformatics mitochondria proteomics redox skeletal muscle 729 730 2 02/02/21 20210201 NES 210201 Intracellular redox homeostasis is crucial for maintaining healthy skeletal muscle. Targeted analytical research has shown that exercise stress induces Nrf2-mediated antioxidant expression in mouse skeletal muscle (Li I et al i . 2015) and, conversely, that Nrf2-deficient mice display higher intramyocellular ROS levels and attenuated antioxidant enzyme abundance (Miller I et al i . 2012). [Extracted from the article]
- Published
- 2021
- Full Text
- View/download PDF
21. Sirt5 regulates chondrocyte metabolism and osteoarthritis development through protein lysine malonylation.
- Author
-
Liu H, Binoy A, Ren S, Martino TC, Miller AE, Willis CRG, Veerabhadraiah SR, Sukul A, Bons J, Rose JP, Schilling B, Jurynec MJ, and Zhu S
- Abstract
Objectives: Chondrocyte metabolic dysfunction plays an important role in osteoarthritis (OA) development during aging and obesity. Protein post-translational modifications (PTMs) have recently emerged as an important regulator of cellular metabolism. We aim to study one type of PTM, lysine malonylation (MaK) and its regulator Sirt5 in OA development., Methods: Human and mouse cartilage tissues were used to measure SIRT5 and MaK levels. Both systemic and cartilage-specific conditional knockout mouse models were subject to high-fat diet (HFD) treatment to induce obesity and OA. Proteomics analysis was performed in Sirt5
-/- and WT chondrocytes. SIRT5 mutation was identified in the Utah Population Database (UPDB)., Results: We found that SIRT5 decreases while MAK increases in the cartilage during aging. A combination of Sirt5 deficiency and obesity exacerbates joint degeneration in a sex dependent manner in mice. We further delineate the malonylome in chondrocytes, pinpointing MaK's predominant impact on various metabolic pathways such as carbon metabolism and glycolysis. Lastly, we identified a rare coding mutation in SIRT5 that dominantly segregates in a family with OA. The mutation results in substitution of an evolutionally invariant phenylalanine (Phe-F) to leucine (Leu-L) (F101L) in the catalytic domain. The mutant protein results in higher MaK level and decreased expression of cartilage ECM genes and upregulation of inflammation associated genes., Conclusions: We found that Sirt5 mediated MaK is an important regulator of chondrocyte cellular metabolism and dysregulation of Sirt5-MaK could be an important mechanism underlying aging and obesity associated OA development.- Published
- 2024
- Full Text
- View/download PDF
22. Adaptability to eccentric exercise training is diminished with age in female mice.
- Author
-
Baumann CW, Deane CS, Etheridge T, Szewczyk NJ, Willis CRG, and Lowe DA
- Subjects
- Female, Animals, Mice, Torque, Muscle, Skeletal physiology
- Abstract
The ability of skeletal muscle to adapt to eccentric contractions has been suggested to be blunted in older muscle. If eccentric exercise is to be a safe and efficient training mode for older adults, preclinical studies need to establish if older muscle can effectively adapt and if not, determine the molecular signatures that are causing this impairment. The purpose of this study was to quantify the extent age impacts functional adaptations of muscle and identify genetic signatures associated with adaptation (or lack thereof). The anterior crural muscles of young (4 mo) and older (28 mo) female mice performed repeated bouts of eccentric contractions in vivo (50 contractions/wk for 5 wk) and isometric torque was measured across the initial and final bouts. Transcriptomics was completed by RNA-sequencing 1 wk following the fifth bout to identify common and differentially regulated genes. When torques post eccentric contractions were compared after the first and fifth bouts, young muscle exhibited a robust ability to adapt, increasing isometric torque 20%-36%, whereas isometric torque of older muscle decreased up to 18% ( P ≤ 0.047). Using differential gene expression, young and older muscles shared some common transcriptional changes in response to eccentric exercise training, whereas other transcripts appeared to be age dependent. That is, the ability to express particular genes after repeated bouts of eccentric contractions was not the same between ages. These molecular signatures may reveal, in part, why older muscles do not appear to be as adaptive to exercise training as young muscles. NEW & NOTEWORTHY The ability to adapt to exercise training may help prevent and combat sarcopenia. Here, we demonstrate young mouse muscles get stronger whereas older mouse muscles become weaker after repeated bouts of eccentric contractions, and that numerous genes were differentially expressed between age groups following training. These results highlight that molecular and functional plasticity is not fixed in skeletal muscle with advancing age, and the ability to handle or cope with physical stress may be impaired.
- Published
- 2023
- Full Text
- View/download PDF
23. Caenorhabditis elegans in microgravity: An omics perspective.
- Author
-
Scott A, Willis CRG, Muratani M, Higashitani A, Etheridge T, Szewczyk NJ, and Deane CS
- Abstract
The application of omics to study Caenorhabditis elegans ( C. elegans ) in the context of spaceflight is increasing, illuminating the wide-ranging biological impacts of spaceflight on physiology. In this review, we highlight the application of omics, including transcriptomics, genomics, proteomics, multi-omics, and integrated omics in the study of spaceflown C. elegans , and discuss the impact, use, and future direction of this branch of research. We highlight the variety of molecular alterations that occur in response to spaceflight, most notably changes in metabolic and neuromuscular gene regulation. These transcriptional features are reproducible and evident across many spaceflown species (e.g., mice and astronauts), supporting the use of C. elegans as a model organism to study spaceflight physiology with translational capital. Integrating tissue-specific, spatial, and multi-omics approaches, which quantitatively link molecular responses to phenotypic adaptations, will facilitate the identification of candidate regulatory molecules for therapeutic intervention and thus represents the next frontiers in C. elegans space omics research., Competing Interests: The authors declare no conflicts of interest., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
24. Routine omics collection is a golden opportunity for European human research in space and analog environments.
- Author
-
Cope H, Willis CRG, MacKay MJ, Rutter LA, Toh LS, Williams PM, Herranz R, Borg J, Bezdan D, Giacomello S, Muratani M, Mason CE, Etheridge T, and Szewczyk NJ
- Abstract
Widespread generation and analysis of omics data have revolutionized molecular medicine on Earth, yet its power to yield new mechanistic insights and improve occupational health during spaceflight is still to be fully realized in humans. Nevertheless, rapid technological advancements and ever-regular spaceflight programs mean that longitudinal, standardized, and cost-effective collection of human space omics data are firmly within reach. Here, we consider the practicality and scientific return of different sampling methods and omic types in the context of human spaceflight. We also appraise ethical and legal considerations pertinent to omics data derived from European astronauts and spaceflight participants (SFPs). Ultimately, we propose that a routine omics collection program in spaceflight and analog environments presents a golden opportunity. Unlocking this bright future of artificial intelligence (AI)-driven analyses and personalized medicine approaches will require further investigation into best practices, including policy design and standardization of omics data, metadata, and sampling methods., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
25. Single-cell transcriptomics identifies master regulators of neurodegeneration in SOD1 ALS iPSC-derived motor neurons.
- Author
-
Namboori SC, Thomas P, Ames R, Hawkins S, Garrett LO, Willis CRG, Rosa A, Stanton LW, and Bhinge A
- Subjects
- Gene Expression Regulation, Gene Regulatory Networks, Humans, Interneurons metabolism, Motor Neurons metabolism, Nerve Degeneration pathology, Signal Transduction, Transforming Growth Factor beta metabolism, Amyotrophic Lateral Sclerosis pathology, Gene Expression Profiling, Induced Pluripotent Stem Cells pathology, Motor Neurons pathology, Nerve Degeneration genetics, Single-Cell Analysis, Superoxide Dismutase-1 metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition characterized by the loss of motor neurons. We utilized single-cell transcriptomics to uncover dysfunctional pathways in degenerating motor neurons differentiated from SOD1 E100G ALS patient-derived induced pluripotent stem cells (iPSCs) and respective isogenic controls. Differential gene expression and network analysis identified activation of developmental pathways and core transcriptional factors driving the ALS motor neuron gene dysregulation. Specifically, we identified activation of SMAD2, a downstream mediator of the transforming growth factor β (TGF-β) signaling pathway as a key driver of SOD1 iPSC-derived motor neuron degeneration. Importantly, our analysis indicates that activation of TGFβ signaling may be a common mechanism shared between SOD1, FUS, C9ORF72, VCP, and sporadic ALS motor neurons. Our results demonstrate the utility of single-cell transcriptomics in mapping disease-relevant gene regulatory networks driving neurodegeneration in ALS motor neurons. We find that ALS-associated mutant SOD1 targets transcriptional networks that perturb motor neuron homeostasis., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. Transcriptomic meta-analysis of disuse muscle atrophy vs. resistance exercise-induced hypertrophy in young and older humans.
- Author
-
Deane CS, Willis CRG, Phillips BE, Atherton PJ, Harries LW, Ames RM, Szewczyk NJ, and Etheridge T
- Subjects
- Aged, Humans, Hypertrophy, Mechanotransduction, Cellular, Muscle, Skeletal, Muscular Atrophy genetics, Transcriptome, Resistance Training
- Abstract
Background: Skeletal muscle atrophy manifests across numerous diseases; however, the extent of similarities/differences in causal mechanisms between atrophying conditions in unclear. Ageing and disuse represent two of the most prevalent and costly atrophic conditions, with resistance exercise training (RET) being the most effective lifestyle countermeasure. We employed gene-level and network-level meta-analyses to contrast transcriptomic signatures of disuse and RET, plus young and older RET to establish a consensus on the molecular features of, and therapeutic targets against, muscle atrophy in conditions of high socio-economic relevance., Methods: Integrated gene-level and network-level meta-analysis was performed on publicly available microarray data sets generated from young (18-35 years) m. vastus lateralis muscle subjected to disuse (unilateral limb immobilization or bed rest) lasting ≥7 days or RET lasting ≥3 weeks, and resistance-trained older (≥60 years) muscle., Results: Disuse and RET displayed predominantly separate transcriptional responses, and transcripts altered across conditions were mostly unidirectional. However, disuse and RET induced directly inverted expression profiles for mitochondrial function and translation regulation genes, with COX4I1, ENDOG, GOT2, MRPL12, and NDUFV2, the central hub components of altered mitochondrial networks, and ZMYND11, a hub gene of altered translation regulation. A substantial number of genes (n = 140) up-regulated post-RET in younger muscle were not similarly up-regulated in older muscle, with young muscle displaying a more pronounced extracellular matrix (ECM) and immune/inflammatory gene expression response. Both young and older muscle exhibited similar RET-induced ubiquitination/RNA processing gene signatures with associated PWP1, PSMB1, and RAF1 hub genes., Conclusions: Despite limited opposing gene profiles, transcriptional signatures of disuse are not simply the converse of RET. Thus, the mechanisms of unloading cannot be derived from studying muscle loading alone and provides a molecular basis for understanding why RET fails to target all transcriptional features of disuse. Loss of RET-induced ECM mechanotransduction and inflammatory profiles might also contribute to suboptimal ageing muscle adaptations to RET. Disuse and age-dependent molecular candidates further establish a framework for understanding and treating disuse/ageing atrophy., (© 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2021
- Full Text
- View/download PDF
27. Comparative Transcriptomics Identifies Neuronal and Metabolic Adaptations to Hypergravity and Microgravity in Caenorhabditis elegans .
- Author
-
Willis CRG, Szewczyk NJ, Costes SV, Udranszky IA, Reinsch SS, Etheridge T, and Conley CA
- Abstract
Deep space exploration is firmly within reach, but health decline during extended spaceflight remains a key challenge. In this study, we performed comparative transcriptomic analysis of Caenorhabditis elegans responses to varying degrees of hypergravity and to two spaceflight experiments (ICE-FIRST and CERISE). We found that progressive hypergravitational load concomitantly increases the extent of differential gene regulation and that subtle changes in ∼1,000 genes are reproducibly observed during spaceflight-induced microgravity. Consequently, we deduce those genes that are concordantly regulated by altered gravity per se or that display inverted expression profiles during hypergravity versus microgravity. Through doing so, we identify several candidate targets with terrestrial roles in neuronal function and/or cellular metabolism, which are linked to regulation by daf-16 /FOXO signaling. These data offer a strong foundation from which to expedite mechanistic understanding of spaceflight-induced maladaptation in higher organisms and, ultimately, promote future targeted therapeutic development., Competing Interests: The authors declare no competing interests., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
28. Network analysis of human muscle adaptation to aging and contraction.
- Author
-
Willis CRG, Ames RM, Deane CS, Phillips BE, Boereboom CL, Abdulla H, Bukhari SSI, Lund JN, Williams JP, Wilkinson DJ, Smith K, Kadi F, Szewczyk NJ, Atherton PJ, and Etheridge T
- Subjects
- Adolescent, Adult, Age Factors, Aged, Aged, 80 and over, Computational Biology methods, Exercise, Female, Gene Expression Profiling, Gene Ontology, Gene Regulatory Networks, Geriatric Assessment, Humans, Male, Transcriptome, Young Adult, Adaptation, Physiological, Aging, Muscle Contraction, Muscle, Skeletal metabolism, Muscle, Skeletal physiopathology
- Abstract
Resistance exercise (RE) remains a primary approach for minimising aging muscle decline. Understanding muscle adaptation to individual contractile components of RE (eccentric, concentric) might optimise RE-based intervention strategies. Herein, we employed a network-driven pipeline to identify putative molecular drivers of muscle aging and contraction mode responses. RNA-sequencing data was generated from young (21±1 y) and older (70±1 y) human skeletal muscle before and following acute unilateral concentric and contralateral eccentric contractions. Application of weighted gene co-expression network analysis identified 33 distinct gene clusters ('modules') with an expression profile regulated by aging, contraction and/or linked to muscle strength. These included two contraction 'responsive' modules (related to 'cell adhesion' and 'transcription factor' processes) that also correlated with the magnitude of post-exercise muscle strength decline. Module searches for 'hub' genes and enriched transcription factor binding sites established a refined set of candidate module-regulatory molecules (536 hub genes and 60 transcription factors) as possible contributors to muscle aging and/or contraction responses. Thus, network-driven analysis can identify new molecular candidates of functional relevance to muscle aging and contraction mode adaptations.
- Published
- 2020
- Full Text
- View/download PDF
29. The acute transcriptional response to resistance exercise: impact of age and contraction mode.
- Author
-
Deane CS, Ames RM, Phillips BE, Weedon MN, Willis CRG, Boereboom C, Abdulla H, Bukhari SSI, Lund JN, Williams JP, Wilkinson DJ, Smith K, Gallagher IJ, Kadi F, Szewczyk NJ, Atherton PJ, and Etheridge T
- Subjects
- Aged, Down-Regulation, Gene Ontology, Healthy Volunteers, Humans, Male, Muscle, Skeletal physiology, RNA-Seq, Transcriptome, Up-Regulation, Young Adult, Aging genetics, Aging physiology, Muscle Contraction genetics, Muscle Contraction physiology, Resistance Training methods
- Abstract
Optimization of resistance exercise (RE) remains a hotbed of research for muscle building and maintenance. However, the interactions between the contractile components of RE (i.e. concentric (CON) and eccentric (ECC)) and age, are poorly defined. We used transcriptomics to compare age-related molecular responses to acute CON and ECC exercise. Eight young (21±1 y) and eight older (70±1 y) exercise-naïve male volunteers had vastus lateralis biopsies collected at baseline and 5 h post unilateral CON and contralateral ECC exercise. RNA was subjected to next-generation sequencing and differentially expressed (DE) genes tested for pathway enrichment using Gene Ontology (GO). The young transcriptional response to CON and ECC was highly similar and older adults displayed moderate contraction-specific profiles, with no GO enrichment. Age-specific responses to ECC revealed 104 DE genes unique to young, and 170 DE genes in older muscle, with no GO enrichment. Following CON, 15 DE genes were young muscle-specific, whereas older muscle uniquely expressed 147 up-regulated genes enriched for cell adhesion and blood vessel development, and 28 down-regulated genes involved in mitochondrial respiration, amino acid and lipid metabolism. Thus, older age is associated with contraction-specific regulation often without clear functional relevance, perhaps reflecting a degree of stochastic age-related dysregulation.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.