1
|
Adamopoulos KI, Sanders LM, Costes SV. NASA GeneLab derived microarray studies of Mus musculus and Homo sapiens organisms in altered gravitational conditions. NPJ Microgravity 2024; 10:49. [PMID: 38671027 PMCID: PMC11053165 DOI: 10.1038/s41526-024-00392-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
One of the greatest challenges of humanity for deep space exploration is to fully understand how altered gravitational conditions affect human physiology. It is evident that the spaceflight environment causes multiple alterations to musculoskeletal, cardiovascular, immune and central nervous systems, to name a few known effects. To better characterize these biological effects, we compare gene expression datasets from microarray studies found in NASA GeneLab, part of the NASA Open Science Data Repository. In this review, we summarize these archived results for various tissues, emphasizing key genes which are highly reproducible in different mice or human experiments. Such exhaustive mining shows the potential of NASA Open Science data to identify and validate mechanisms taking place when mammalian organisms are exposed to microgravity or other spaceflight conditions. Our comparative meta-analysis findings highlight certain degrees of overlap and reproducibility in genes identified as differentially expressed within musculoskeletal tissues in each species across a variety of altered gravity conditions. However, the level of overlap between species was found to be significantly limited, partly attributed to the limited availability of human samples.
Collapse
Affiliation(s)
- Konstantinos I Adamopoulos
- National Technical University of Athens, School of Electrical and Computer Engineering, Biomedical Engineering Laboratory, Zografou, Athens, Greece
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Lauren M Sanders
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- NASA Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sylvain V Costes
- NASA Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
2
|
Takahashi A, Honda Y, Tanaka N, Miyake J, Maeda S, Kataoka H, Sakamoto J, Okita M. Skeletal Muscle Electrical Stimulation Prevents Progression of Disuse Muscle Atrophy via Forkhead Box O Dynamics Mediated by Phosphorylated Protein Kinase B and Peroxisome Proliferator-Activated Receptor gamma Coactivator-1alpha. Physiol Res 2024; 73:105-115. [PMID: 38466009 PMCID: PMC11019614 DOI: 10.33549/physiolres.935157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/12/2023] [Indexed: 04/26/2024] Open
Abstract
Although electrical muscle stimulation (EMS) of skeletal muscle effectively prevents muscle atrophy, its effect on the breakdown of muscle component proteins is unknown. In this study, we investigated the biological mechanisms by which EMS-induced muscle contraction inhibits disuse muscle atrophy progression. Experimental animals were divided into a control group and three experimental groups: immobilized (Im; immobilization treatment), low-frequency (LF; immobilization treatment and low-frequency muscle contraction exercise), and high-frequency (HF; immobilization treatment and high-frequency muscle contraction exercise). Following the experimental period, bilateral soleus muscles were collected and analyzed. Atrogin-1 and Muscle RING finger 1 (MuRF-1) mRNA expression levels were significantly higher for the experimental groups than for the control group but were significantly lower for the HF group than for the Im group. Peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) mRNA and protein expression levels in the HF group were significantly higher than those in the Im group, with no significant differences compared to the Con group. Both the Forkhead box O (FoxO)/phosphorylated FoxO and protein kinase B (AKT)/phosphorylated AKT ratios were significantly lower for the Im group than for the control group and significantly higher for the HF group than for the Im group. These results, the suppression of atrogin-1 and MuRF-1 expression for the HF group may be due to decreased nuclear expression of FoxO by AKT phosphorylation and suppression of FoxO transcriptional activity by PGC-1alpha. Furthermore, the number of muscle contractions might be important for effective EMS.
Collapse
Affiliation(s)
- A Takahashi
- Institute of Biomedical Sciences (Health Sciences), Nagasaki University, Nagasaki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Duscher AA, Vroom MM, Foster JS. Impact of modeled microgravity stress on innate immunity in a beneficial animal-microbe symbiosis. Sci Rep 2024; 14:2912. [PMID: 38316910 PMCID: PMC10844198 DOI: 10.1038/s41598-024-53477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
The innate immune response is the first line of defense for all animals to not only detect invading microbes and toxins but also sense and interface with the environment. One such environment that can significantly affect innate immunity is spaceflight. In this study, we explored the impact of microgravity stress on key elements of the NFκB innate immune pathway. The symbiosis between the bobtail squid Euprymna scolopes and its beneficial symbiont Vibrio fischeri was used as a model system under a simulated microgravity environment. The expression of genes associated with the NFκB pathway was monitored over time as the symbiosis progressed. Results revealed that although the onset of the symbiosis was the major driver in the differential expression of NFκB signaling, the stress of simulated low-shear microgravity also caused a dysregulation of expression. Several genes were expressed at earlier time points suggesting that elements of the E. scolopes NFκB pathway are stress-inducible, whereas expression of other pathway components was delayed. The results provide new insights into the role of NFκB signaling in the squid-vibrio symbiosis, and how the stress of microgravity negatively impacts the host immune response. Together, these results provide a foundation to develop mitigation strategies to maintain host-microbe homeostasis during spaceflight.
Collapse
Affiliation(s)
- Alexandrea A Duscher
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Chesapeake Bay Governor's School, Warsaw, VA, 22572, USA
| | - Madeline M Vroom
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Vaxxinity, Space Life Sciences Lab, Merritt Island, FL, 32953, USA
| | - Jamie S Foster
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA.
| |
Collapse
|
4
|
Dickerson BL, Sowinski R, Kreider RB, Wu G. Impacts of microgravity on amino acid metabolism during spaceflight. Exp Biol Med (Maywood) 2023; 248:380-393. [PMID: 36775855 PMCID: PMC10281620 DOI: 10.1177/15353702221139189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
Spaceflight exerts an extreme and unique influence on human physiology as astronauts are subjected to long-term or short-term exposure to microgravity. During spaceflight, a multitude of physiological changes, including the loss of skeletal muscle mass, bone resorption, oxidative stress, and impaired blood flow, occur, which can affect astronaut health and the likelihood of mission success. In vivo and in vitro metabolite studies suggest that amino acids are among the most affected nutrients and metabolites by microgravity (a weightless condition due to very weak gravitational forces). Moreover, exposure to microgravity alters gut microbial composition, immune function, musculoskeletal health, and consequently amino acid metabolism. Appropriate knowledge of daily protein consumption, with a focus on specific functional amino acids, may offer insight into potential combative and/or therapeutic effects of amino acid consumption in astronauts and space travelers. This will further aid in the successful development of long-term manned space mission and permanent space habitats.
Collapse
Affiliation(s)
- Broderick L Dickerson
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Ryan Sowinski
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Richard B Kreider
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Guoyao Wu
- Department of Animal Science and
Faculty of Nutrition, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Malhan D, Yalçin M, Schoenrock B, Blottner D, Relógio A. Skeletal muscle gene expression dysregulation in long-term spaceflights and aging is clock-dependent. NPJ Microgravity 2023; 9:30. [PMID: 37012297 PMCID: PMC10070655 DOI: 10.1038/s41526-023-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/13/2023] [Indexed: 04/05/2023] Open
Abstract
The circadian clock regulates cellular and molecular processes in mammals across all tissues including skeletal muscle, one of the largest organs in the human body. Dysregulated circadian rhythms are characteristic of aging and crewed spaceflight, associated with, for example, musculoskeletal atrophy. Molecular insights into spaceflight-related alterations of circadian regulation in skeletal muscle are still missing. Here, we investigated potential functional consequences of clock disruptions on skeletal muscle using published omics datasets obtained from spaceflights and other clock-altering, external (fasting and exercise), or internal (aging) conditions on Earth. Our analysis identified alterations of the clock network and skeletal muscle-associated pathways, as a result of spaceflight duration in mice, which resembles aging-related gene expression changes observed in humans on Earth (e.g., ATF4 downregulation, associated with muscle atrophy). Furthermore, according to our results, external factors such as exercise or fasting lead to molecular changes in the core-clock network, which may compensate for the circadian disruption observed during spaceflights. Thus, maintaining circadian functioning is crucial to ameliorate unphysiological alterations and musculoskeletal atrophy reported among astronauts.
Collapse
Affiliation(s)
- Deeksha Malhan
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany
| | - Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany
| | - Britt Schoenrock
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
| | - Dieter Blottner
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Neuromuscular System and Neuromuscular Signaling, Berlin Center of Space Medicine & Extreme Environments, Berlin, 10115, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, 20457, Germany.
| |
Collapse
|
6
|
Capri M, Conte M, Ciurca E, Pirazzini C, Garagnani P, Santoro A, Longo F, Salvioli S, Lau P, Moeller R, Jordan J, Illig T, Villanueva MM, Gruber M, Bürkle A, Franceschi C, Rittweger J. Long-term human spaceflight and inflammaging: Does it promote aging? Ageing Res Rev 2023; 87:101909. [PMID: 36918115 DOI: 10.1016/j.arr.2023.101909] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Spaceflight and its associated stressors, such as microgravity, radiation exposure, confinement, circadian derailment and disruptive workloads represent an unprecedented type of exposome that is entirely novel from an evolutionary stand point. Within this perspective, we aimed to review the effects of prolonged spaceflight on immune-neuroendocrine systems, brain and brain-gut axis, cardiovascular system and musculoskeletal apparatus, highlighting in particular the similarities with an accelerated aging process. In particular, spaceflight-induced muscle atrophy/sarcopenia and bone loss, vascular and metabolic changes, hyper and hypo reaction of innate and adaptive immune system appear to be modifications shared with the aging process. Most of these modifications are mediated by molecular events that include oxidative and mitochondrial stress, autophagy, DNA damage repair and telomere length alteration, among others, which directly or indirectly converge on the activation of an inflammatory response. According to the inflammaging theory of aging, such an inflammatory response could be a driver of an acceleration of the normal, physiological rate of aging and it is likely that all the systemic modifications in turn lead to an increase of inflammaging in a sort of vicious cycle. The most updated countermeasures to fight these modifications will be also discussed in the light of their possible application not only for astronauts' benefit, but also for older adults on the ground.
Collapse
Affiliation(s)
- Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.
| | - Erika Ciurca
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Chiara Pirazzini
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy; Clinical Chemistry Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden; CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy; Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Aurelia Santoro
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Federica Longo
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Patrick Lau
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Ralf Moeller
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Thomas Illig
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Maria-Moreno Villanueva
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Markus Gruber
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Alexander Bürkle
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Claudio Franceschi
- Department of Applied Mathematics of the Institute of ITMM, National Research Lobachevsky State University of Nizhny Novgorod, the Russian Federation
| | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
7
|
Sanesi L, Storlino G, Dicarlo M, Oranger A, Zerlotin R, Pignataro P, Suriano C, Guida G, Grano M, Colaianni G, Colucci SC. Time-dependent unloading effects on muscle and bone and involvement of FNDC5/irisin axis. NPJ Microgravity 2023; 9:4. [PMID: 36658231 PMCID: PMC9852594 DOI: 10.1038/s41526-023-00251-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
The identification of biomarkers and countermeasures to prevent the adverse effects on the musculoskeletal system caused by the absence of mechanical loading is the main goal of space biomedical research studies. In this study, we analyzed over 4 weeks of unloading, the modulation in the expression of key proteins in Vastus lateralis, Gastrocnemius and cortical bone in parallel with the modulation of irisin serum levels and its precursor FNDC5 in skeletal muscle of hind limb unloaded (HU) mice. Here we report that Atrogin-1 was up-regulated as early as 1- and 2-week of unloading, whereas Murf-1 at 2- and 3-weeks, along with a marked modulation in the expression of myosin heavy chain isoforms during unloading. Since HU mice showed reduced irisin serum levels at 4-weeks, as well as FNDC5 decrease at 3- and 4-weeks, we treated HU mice with recombinant irisin for 4 weeks, showing that unloading-dependent decline of myosin heavy chain isoforms, MyHCIIα and MyHCIIx, and the anti-apoptotic factor Bcl2, were prevented. In parallel, irisin treatment inhibited the increase of the senescence marker p53, and the pro-apoptotic factor Bax. Overall, these results suggest that the myokine irisin could be a possible therapy to counteract the musculoskeletal impairment caused by unloading.
Collapse
Affiliation(s)
- Lorenzo Sanesi
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Manuela Dicarlo
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Angela Oranger
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Roberta Zerlotin
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Patrizia Pignataro
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy ,grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Clelia Suriano
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Gabriella Guida
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| | - Maria Grano
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Graziana Colaianni
- grid.7644.10000 0001 0120 3326Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Silvia Concetta Colucci
- grid.7644.10000 0001 0120 3326Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy
| |
Collapse
|
8
|
Hanson AM, Young MH, Harrison BC, Zhou X, Han HQ, Stodieck LS, Ferguson VL. Inhibiting myostatin signaling partially mitigates structural and functional adaptations to hindlimb suspension in mice. NPJ Microgravity 2023; 9:2. [PMID: 36646717 PMCID: PMC9842652 DOI: 10.1038/s41526-022-00233-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/12/2022] [Indexed: 01/18/2023] Open
Abstract
Novel treatments for muscle wasting are of significant value to patients with disease states that result in muscle weakness, injury recovery after immobilization and bed rest, and for astronauts participating in long-duration spaceflight. We utilized an anti-myostatin peptibody to evaluate how myostatin signaling contributes to muscle loss in hindlimb suspension. Male C57BL/6 mice were left non-suspended (NS) or were hindlimb suspended (HS) for 14 days and treated with a placebo vehicle (P) or anti-myostatin peptibody (D). Hindlimb suspension (HS-P) resulted in rapid and significantly decreased body mass (-5.6% by day 13) with hindlimb skeletal muscle mass losses between -11.2% and -22.5% and treatment with myostatin inhibitor (HS-D) partially attenuated these losses. Myostatin inhibition increased hindlimb strength with no effect on soleus tetanic strength. Soleus mass and fiber CSA were reduced with suspension and did not increase with myostatin inhibition. In contrast, the gastrocnemius showed histological evidence of wasting with suspension that was partially mitigated with myostatin inhibition. While expression of genes related to protein degradation (Atrogin-1 and Murf-1) in the tibialis anterior increased with suspension, these atrogenes were not significantly reduced by myostatin inhibition despite a modest activation of the Akt/mTOR pathway. Taken together, these findings suggest that myostatin is important in hindlimb suspension but also motivates the study of other factors that contribute to disuse muscle wasting. Myostatin inhibition benefitted skeletal muscle size and function, which suggests therapeutic potential for both spaceflight and terrestrial applications.
Collapse
Affiliation(s)
- Andrea M. Hanson
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Mary H. Young
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Brooke C. Harrison
- grid.266190.a0000000096214564Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO USA
| | - Xiaolan Zhou
- grid.417886.40000 0001 0657 5612Amgen Inc., Thousand Oaks, CA USA ,Present Address: AliveGen USA Inc., Thousand Oaks, CA USA
| | - H. Q. Han
- grid.417886.40000 0001 0657 5612Amgen Inc., Thousand Oaks, CA USA ,Present Address: AliveGen USA Inc., Thousand Oaks, CA USA
| | - Louis S. Stodieck
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Virginia L. Ferguson
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA ,grid.266190.a0000000096214564Department of Mechanical Engineering, University of Colorado, Boulder, CO USA ,grid.266190.a0000000096214564BioFrontiers Institute, University of Colorado, Boulder, CO USA
| |
Collapse
|
9
|
Are Skeletal Muscle Changes during Prolonged Space Flights Similar to Those Experienced by Frail and Sarcopenic Older Adults? LIFE (BASEL, SWITZERLAND) 2022; 12:life12122139. [PMID: 36556504 PMCID: PMC9781047 DOI: 10.3390/life12122139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Microgravity exposure causes several physiological and psychosocial alterations that challenge astronauts' health during space flight. Notably, many of these changes are mostly related to physical inactivity influencing different functional systems and organ biology, in particular the musculoskeletal system, dramatically resulting in aging-like phenotypes, such as those occurring in older persons on Earth. In this sense, sarcopenia, a syndrome characterized by the loss in muscle mass and strength due to skeletal muscle unloading, is undoubtedly one of the most critical aging-like adverse effects of microgravity and a prevalent problem in the geriatric population, still awaiting effective countermeasures. Therefore, there is an urgent demand to identify clinically relevant biological markers and to underline molecular mechanisms behind these effects that are still poorly understood. From this perspective, a lesson from Geroscience may help tailor interventions to counteract the adverse effects of microgravity. For instance, decades of studies in the field have demonstrated that in the older people, the clinical picture of sarcopenia remarkably overlaps (from a clinical and biological point of view) with that of frailty, primarily when referred to the physical function domain. Based on this premise, here we provide a deeper understanding of the biological mechanisms of sarcopenia and frailty, which in aging are often considered together, and how these converge with those observed in astronauts after space flight.
Collapse
|
10
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
11
|
Wang S, Fang L, Cong L, Chung JPW, Li TC, Chan DYL. Myostatin: a multifunctional role in human female reproduction and fertility - a short review. Reprod Biol Endocrinol 2022; 20:96. [PMID: 35780124 PMCID: PMC9250276 DOI: 10.1186/s12958-022-00969-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/21/2022] [Indexed: 11/10/2022] Open
Abstract
Myostatin (MSTN) is member of the transforming growth factor β (TGF-β) superfamily and was originally identified in the musculoskeletal system as a negative regulator of skeletal muscle growth. The functional roles of MSTN outside of the musculoskeletal system have aroused researchers' interest in recent years, with an increasing number of studies being conducted in this area. Notably, the expression of MSTN and its potential activities in various reproductive organs, including the ovary, placenta, and uterus, have recently been examined. Numerous studies published in the last few years demonstrate that MSTN plays a critical role in human reproduction and fertility, including the regulation of follicular development, ovarian steroidogenesis, granule-cell proliferation, and oocyte maturation regulation. Furthermore, findings from clinical samples suggest that MSTN may play a key role in the pathogenesis of several reproductive disorders such as uterine myoma, preeclampsia (PE), ovary hyperstimulation syndrome (OHSS), and polycystic ovarian syndrome (PCOS). There is no comprehensive review regarding to MSTN related to the female reproductive system in the literature. This review serves as a summary of the genes in reproductive medicine and their potential influence. We summarized MSTN expression in different compartments of the female reproductive system. Subsequently, we discuss the role of MSTN in both physiological and several pathological conditions related to the female fertility and reproduction-related diseases.
Collapse
Affiliation(s)
- Sijia Wang
- Assisted reproductive technologies unit, Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, SAR, China
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Luping Cong
- Assisted reproductive technologies unit, Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, SAR, China
| | - Jacqueline Pui Wah Chung
- Assisted reproductive technologies unit, Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, SAR, China
| | - Tin Chiu Li
- Assisted reproductive technologies unit, Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, SAR, China
| | - David Yiu Leung Chan
- Assisted reproductive technologies unit, Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, SAR, China.
| |
Collapse
|
12
|
Barbé C, Salles J, Chambon C, Giraudet C, Sanchez P, Patrac V, Denis P, Boirie Y, Walrand S, Gueugneau M. Characterization of the Skeletal Muscle Proteome in Undernourished Old Rats. Int J Mol Sci 2022; 23:ijms23094762. [PMID: 35563153 PMCID: PMC9101871 DOI: 10.3390/ijms23094762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Aging is associated with a progressive loss of skeletal muscle mass and function termed sarcopenia. Various metabolic alterations that occur with aging also increase the risk of undernutrition, which can worsen age-related sarcopenia. However, the impact of undernutrition on aged skeletal muscle remains largely under-researched. To build a deeper understanding of the cellular and molecular mechanisms underlying age-related sarcopenia, we characterized the undernutrition-induced changes in the skeletal muscle proteome in old rats. For this study, 20-month-old male rats were fed 50% or 100% of their spontaneous intake for 12 weeks, and proteomic analysis was performed on both slow- and fast-twitch muscles. Proteomic profiling of undernourished aged skeletal muscle revealed that undernutrition has profound effects on muscle proteome independently of its effect on muscle mass. Undernutrition-induced changes in muscle proteome appear to be muscle-type-specific: slow-twitch muscle showed a broad pattern of differential expression in proteins important for energy metabolism, whereas fast-twitch muscle mainly showed changes in protein turnover between undernourished and control rats. This first proteomic analysis of undernourished aged skeletal muscle provides new molecular-level insight to explain phenotypic changes in undernourished aged muscle. We anticipate this work as a starting point to define new biomarkers associated with undernutrition-induced muscle loss in the elderly.
Collapse
Affiliation(s)
- Caroline Barbé
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Jérôme Salles
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Christophe Chambon
- Animal Products Quality Unit (QuaPA), INRAE, 63122 Clermont-Ferrand, France;
- Metabolomic and Proteomic Exploration Facility, Clermont Auvergne University, INRAE, 63122 Clermont-Ferrand, France
| | - Christophe Giraudet
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Phelipe Sanchez
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Véronique Patrac
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Philippe Denis
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
| | - Yves Boirie
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
- Department of Clinical Nutrition, Clermont-Ferrand University Hospital Center, 63000 Clermont-Ferrand, France
| | - Stéphane Walrand
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
- Department of Clinical Nutrition, Clermont-Ferrand University Hospital Center, 63000 Clermont-Ferrand, France
| | - Marine Gueugneau
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, 63000 Clermont-Ferrand, France; (C.B.); (J.S.); (C.G.); (P.S.); (V.P.); (P.D.); (Y.B.); (S.W.)
- Correspondence: ; Tel.: +33-4-73-60-82-65
| |
Collapse
|
13
|
Detection of Target Genes for Drug Repurposing to Treat Skeletal Muscle Atrophy in Mice Flown in Spaceflight. Genes (Basel) 2022; 13:genes13030473. [PMID: 35328027 PMCID: PMC8953707 DOI: 10.3390/genes13030473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/25/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle atrophy is a common condition in aging, diabetes, and in long duration spaceflights due to microgravity. This article investigates multi-modal gene disease and disease drug networks via link prediction algorithms to select drugs for repurposing to treat skeletal muscle atrophy. Key target genes that cause muscle atrophy in the left and right extensor digitorum longus muscle tissue, gastrocnemius, quadriceps, and the left and right soleus muscles are detected using graph theoretic network analysis, by mining the transcriptomic datasets collected from mice flown in spaceflight made available by GeneLab. We identified the top muscle atrophy gene regulators by the Pearson correlation and Bayesian Markov blanket method. The gene disease knowledge graph was constructed using the scalable precision medicine knowledge engine. We computed node embeddings, random walk measures from the networks. Graph convolutional networks, graph neural networks, random forest, and gradient boosting methods were trained using the embeddings, network features for predicting links and ranking top gene-disease associations for skeletal muscle atrophy. Drugs were selected and a disease drug knowledge graph was constructed. Link prediction methods were applied to the disease drug networks to identify top ranked drugs for therapeutic treatment of skeletal muscle atrophy. The graph convolution network performs best in link prediction based on receiver operating characteristic curves and prediction accuracies. The key genes involved in skeletal muscle atrophy are associated with metabolic and neurodegenerative diseases. The drugs selected for repurposing using the graph convolution network method were nutrients, corticosteroids, anti-inflammatory medications, and others related to insulin.
Collapse
|
14
|
Fast and slow myofiber-specific expression profiles are affected by noncoding RNAs in Mongolian horses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2022; 41:100942. [PMID: 34823143 DOI: 10.1016/j.cbd.2021.100942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
The heterogeneity and plasticity of muscle fibers are essential for the athletic performance of horses, mainly at the adaption of exercises and the effect on muscle diseases. Skeletal muscle fibers can be generally distinguished by their characteristics of contraction as slow and fast type myofibers. The diversity of contractile properties and metabolism enable skeletal muscles to respond to the variable functional requirements. We investigated the muscle fiber composition and metabolic enzyme activities of splenius muscle and gluteus medius muscle from Mongolian horses. The deep RNA-seq analysis of detecting differentially expressed mRNAs, lncRNAs, circRNAs and their correlation analysis from two muscles were performed. Splenius muscle and gluteus medius muscle from Mongolian horses showed a high divergence of myofiber compositions and metabolic enzyme activities. Corresponding to their phenotypic characteristics, 57 differentially expressed long noncoding RNAs and 12 differentially expressed circle RNAs were found between two muscles. The analysis results indicate multiple binding sites were detected in lncRNAs and circRNAs with myofiber-specific expressed miRNAs. Among which we found significant correlations between the above noncoding RNAs, miRNAs, their target genes, myofiber-specific developmental transcript factors, and sarcomere genes. We suggest that the ceRNA mechanism of differentially expressed noncoding RNAs by acting as miRNA sponges could be fine tuners in regulating skeletal muscle fiber composition and transition in horses, which will operate new protective measures of muscle disease and locomotor adaption for racehorses.
Collapse
|
15
|
Tran KN, Choi JI. Mimic microgravity effect on muscle transcriptome under ionizing radiation. LIFE SCIENCES IN SPACE RESEARCH 2022; 32:96-104. [PMID: 35065767 DOI: 10.1016/j.lssr.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Spaceflight imposes the risk of skeletal muscle atrophy for astronauts. Two main factors of a spaceflight that results in deleterious effects are microgravity and cosmic rays in outer space. To study spaceflight-induced muscle atrophy with ground-based models, we performed two models of microgravity, tail suspension and denervation, in a low dose radiation environment and studied transcriptional changes in rat soleus muscle using microarrays. Soleus muscle from rats in the denervation group had greater expression changes compared to that found in rats from the tail suspension group. However, there was a very similar pattern of expression of differentially expressed genes (DEGs) in both models. In total, we identified 144 differentially expressed genes common in both models. Our study yielded two main findings. First, a large number of genes involved in energy metabolism were transcriptionally suppressed including those involved in fatty acid transport and beta-oxidation, and oxidative phosphorylation. Second, slow-twitch contractile protein encoding genes were down-regulated while there was an up-regulation in the fast-twitch type transcription. These results were consistent with other spaceflight studies on the effects on muscle cells, hence showed the potential of our ground-based models in studying spaceflight effects. The genes that might be involved in spaceflight effects will serve as candidate genes for future studies in understanding the mechanism of spaceflight-induced muscle atrophy and result in the development of effective countermeasures.
Collapse
Affiliation(s)
- Kim Ngan Tran
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jong-Il Choi
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
16
|
Esposito P, Picciotto D, Battaglia Y, Costigliolo F, Viazzi F, Verzola D. Myostatin: Basic biology to clinical application. Adv Clin Chem 2022; 106:181-234. [PMID: 35152972 DOI: 10.1016/bs.acc.2021.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myostatin is a member of the transforming growth factor (TGF)-β superfamily. It is expressed by animal and human skeletal muscle cells where it limits muscle growth and promotes protein breakdown. Its effects are influenced by complex mechanisms including transcriptional and epigenetic regulation and modulation by extracellular binding proteins. Due to its actions in promoting muscle atrophy and cachexia, myostatin has been investigated as a promising therapeutic target to counteract muscle mass loss in experimental models and patients affected by different muscle-wasting conditions. Moreover, growing evidence indicates that myostatin, beyond to regulate skeletal muscle growth, may have a role in many physiologic and pathologic processes, such as obesity, insulin resistance, cardiovascular and chronic kidney disease. In this chapter, we review myostatin biology, including intracellular and extracellular regulatory pathways, and the role of myostatin in modulating physiologic processes, such as muscle growth and aging. Moreover, we discuss the most relevant experimental and clinical evidence supporting the extra-muscle effects of myostatin. Finally, we consider the main strategies developed and tested to inhibit myostatin in clinical trials and discuss the limits and future perspectives of the research on myostatin.
Collapse
Affiliation(s)
- Pasquale Esposito
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Daniela Picciotto
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Yuri Battaglia
- Nephrology and Dialysis Unit, St. Anna University Hospital, Ferrara, Italy
| | - Francesca Costigliolo
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
17
|
Sharlo K, Tyganov SA, Tomilovskaya E, Popov DV, Saveko AA, Shenkman BS. Effects of Various Muscle Disuse States and Countermeasures on Muscle Molecular Signaling. Int J Mol Sci 2021; 23:ijms23010468. [PMID: 35008893 PMCID: PMC8745071 DOI: 10.3390/ijms23010468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle is capable of changing its structural parameters, metabolic rate and functional characteristics within a wide range when adapting to various loading regimens and states of the organism. Prolonged muscle inactivation leads to serious negative consequences that affect the quality of life and work capacity of people. This review examines various conditions that lead to decreased levels of muscle loading and activity and describes the key molecular mechanisms of muscle responses to these conditions. It also details the theoretical foundations of various methods preventing adverse muscle changes caused by decreased motor activity and describes these methods. A number of recent studies presented in this review make it possible to determine the molecular basis of the countermeasure methods used in rehabilitation and space medicine for many years, as well as to identify promising new approaches to rehabilitation and to form a holistic understanding of the mechanisms of gravity force control over the muscular system.
Collapse
|
18
|
Srivastava S, Rathor R, Singh SN, Suryakumar G. Emerging role of MyomiRs as biomarkers and therapeutic targets in skeletal muscle diseases. Am J Physiol Cell Physiol 2021; 321:C859-C875. [PMID: 34586896 DOI: 10.1152/ajpcell.00057.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several chronic diseases lead to skeletal muscle loss and a decline in physical performance. MicroRNAs (miRNAs) are small, noncoding RNAs, which have exhibited their role in the development and diseased state of the skeletal muscle. miRNA regulates gene expression by binding to the 3' untranslated region of its target mRNA. Due to the robust stability in biological fluids, miRNAs are ideal candidate as biomarker. These miRNAs provide a novel avenue in strengthening our awareness and knowledge about the factors governing skeletal muscle functions such as development, growth, metabolism, differentiation, and cell proliferation. It also helps in understanding the therapeutic strategies in improving or conserving skeletal muscle health. This review outlines the evidence regarding the present knowledge on the role miRNA as a potential biomarker in skeletal muscle diseases and their exploration might be a unique and potential therapeutic strategy for various skeletal muscle disorders.
Collapse
Affiliation(s)
| | - Richa Rathor
- Defence Institute of Physiology & Allied Sciences (DIPAS), Delhi, India
| | - Som Nath Singh
- Defence Institute of Physiology & Allied Sciences (DIPAS), Delhi, India
| | - Geetha Suryakumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Delhi, India
| |
Collapse
|
19
|
Wilburn D, Ismaeel A, Machek S, Fletcher E, Koutakis P. Shared and distinct mechanisms of skeletal muscle atrophy: A narrative review. Ageing Res Rev 2021; 71:101463. [PMID: 34534682 DOI: 10.1016/j.arr.2021.101463] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022]
Abstract
Maintenance of skeletal muscle mass and function is an incredibly nuanced balance of anabolism and catabolism that can become distorted within different pathological conditions. In this paper we intend to discuss the distinct intracellular signaling events that regulate muscle protein atrophy for a given clinical occurrence. Aside from the common outcome of muscle deterioration, several conditions have at least one or more distinct mechanisms that creates unique intracellular environments that facilitate muscle loss. The subtle individuality to each of these given pathologies can provide both researchers and clinicians with specific targets of interest to further identify and increase the efficacy of medical treatments and interventions.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76706, USA
| | - Steven Machek
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Emma Fletcher
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; Department of Biology, Baylor University, Waco, TX 76706, USA
| | | |
Collapse
|
20
|
Ohira T, Ino Y, Kimura Y, Nakai Y, Kimura A, Kurata Y, Kagawa H, Kimura M, Egashira K, Matsuda C, Ohira Y, Furukawa S, Hirano H. Effects of microgravity exposure and fructo-oligosaccharide ingestion on the proteome of soleus and extensor digitorum longus muscles in developing mice. NPJ Microgravity 2021; 7:34. [PMID: 34535681 PMCID: PMC8448765 DOI: 10.1038/s41526-021-00164-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Short-chain fatty acids produced by the gut bacterial fermentation of non-digestible carbohydrates, e.g., fructo-oligosaccharide (FOS), contribute to the maintenance of skeletal muscle mass and oxidative metabolic capacity. We evaluated the effect of FOS ingestion on protein expression of soleus (Sol) and extensor digitorum longus muscles in mice exposed to microgravity (μ-g). Twelve 9-week-old male C57BL/6J mice were raised individually on the International Space Station under μ-g or artificial 1-g and fed a diet with or without FOS (n = 3/group). Regardless of FOS ingestion, the absolute wet weights of both muscles tended to decrease, and the fiber phenotype in Sol muscles shifted toward fast-twitch type following μ-g exposure. However, FOS ingestion tended to mitigate the μ-g-exposure-related decrease in oxidative metabolism and enhance glutathione redox detoxification in Sol muscles. These results indicate that FOS ingestion mildly suppresses metabolic changes and oxidative stress in antigravity Sol muscles during spaceflight.
Collapse
Affiliation(s)
- Takashi Ohira
- Research Center for Space and Medical Sciences and Organization for Research Initiatives and Development, Doshisha University, Kyoto, Japan. .,Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi Osaka-Sayama, Osaka, Japan. .,Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan. .,Space Biomedical Research Group, Japan Aerospace Exploration Agency, Ibaraki, Japan.
| | - Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Ayuko Kimura
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Yoichi Kurata
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Hiroyuki Kagawa
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Mitsuo Kimura
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Kenji Egashira
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Chie Matsuda
- Space Biomedical Research Group, Japan Aerospace Exploration Agency, Ibaraki, Japan
| | - Yoshinobu Ohira
- Research Center for Space and Medical Sciences and Organization for Research Initiatives and Development, Doshisha University, Kyoto, Japan
| | - Satoshi Furukawa
- Space Biomedical Research Group, Japan Aerospace Exploration Agency, Ibaraki, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| |
Collapse
|
21
|
Cahill T, Cope H, Bass JJ, Overbey EG, Gilbert R, da Silveira WA, Paul AM, Mishra T, Herranz R, Reinsch SS, Costes SV, Hardiman G, Szewczyk NJ, Tahimic CGT. Mammalian and Invertebrate Models as Complementary Tools for Gaining Mechanistic Insight on Muscle Responses to Spaceflight. Int J Mol Sci 2021; 22:ijms22179470. [PMID: 34502375 PMCID: PMC8430797 DOI: 10.3390/ijms22179470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Bioinformatics approaches have proven useful in understanding biological responses to spaceflight. Spaceflight experiments remain resource intensive and rare. One outstanding issue is how to maximize scientific output from a limited number of omics datasets from traditional animal models including nematodes, fruitfly, and rodents. The utility of omics data from invertebrate models in anticipating mammalian responses to spaceflight has not been fully explored. Hence, we performed comparative analyses of transcriptomes of soleus and extensor digitorum longus (EDL) in mice that underwent 37 days of spaceflight. Results indicate shared stress responses and altered circadian rhythm. EDL showed more robust growth signals and Pde2a downregulation, possibly underlying its resistance to atrophy versus soleus. Spaceflight and hindlimb unloading mice shared differential regulation of proliferation, circadian, and neuronal signaling. Shared gene regulation in muscles of humans on bedrest and space flown rodents suggest targets for mitigating muscle atrophy in space and on Earth. Spaceflight responses of C. elegans were more similar to EDL. Discrete life stages of D. melanogaster have distinct utility in anticipating EDL and soleus responses. In summary, spaceflight leads to shared and discrete molecular responses between muscle types and invertebrate models may augment mechanistic knowledge gained from rodent spaceflight and ground-based studies.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
| | - Henry Cope
- Nottingham Biomedical Research Centre (BRC), School of Computer Science, University of Nottingham, Nottingham NG7 2QL, UK;
| | - Joseph J. Bass
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), University of Nottingham, Nottingham NG7 2QL, UK; (J.J.B.); (N.J.S.)
| | - Eliah G. Overbey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Rachel Gilbert
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Universities Space Research Association, Columbia, MD 21046, USA
| | - Willian Abraham da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
- Department of Biological Sciences, School of Life Sciences and Education, Staffordshire University, Stoke-on-Trent ST4 2DF, UK
| | - Amber M. Paul
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL 32114, USA
- Blue Marble Space Institute of Science, Seattle, WA 98104, USA
| | - Tejaswini Mishra
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA 94305, USA;
| | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas–CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain;
| | - Sigrid S. Reinsch
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.); (G.H.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathaniel J. Szewczyk
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), University of Nottingham, Nottingham NG7 2QL, UK; (J.J.B.); (N.J.S.)
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Candice G. T. Tahimic
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA; (R.G.); (A.M.P.); (S.S.R.); (S.V.C.)
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
- Correspondence:
| |
Collapse
|
22
|
Liang Y, Wang M, Liu Y, Wang C, Takahashi K, Naruse K. Meta-Analysis-Assisted Detection of Gravity-Sensitive Genes in Human Vascular Endothelial Cells. Front Cell Dev Biol 2021; 9:689662. [PMID: 34422812 PMCID: PMC8371407 DOI: 10.3389/fcell.2021.689662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Gravity affects the function and maintenance of organs, such as bones, muscles, and the heart. Several studies have used DNA microarrays to identify genes with altered expressions in response to gravity. However, it is technically challenging to combine the results from various microarray datasets because of their different data structures. We hypothesized that it is possible to identify common changes in gene expression from the DNA microarray datasets obtained under various conditions and methods. In this study, we grouped homologous genes to perform a meta-analysis of multiple vascular endothelial cell and skeletal muscle datasets. According to the t-distributed stochastic neighbor embedding (t-SNE) analysis, the changes in the gene expression pattern in vascular endothelial cells formed specific clusters. We also identified candidate genes in endothelial cells that responded to gravity. Further, we exposed human umbilical vein endothelial cells (HUVEC) to simulated microgravity (SMG) using a clinostat and measured the expression levels of the candidate genes. Gene expression analysis using qRT-PCR revealed that the expression level of the prostaglandin (PG) transporter gene SLCO2A1 decreased in response to microgravity, consistent with the meta-analysis of microarray datasets. Furthermore, the direction of gravity affected the expression level of SLCO2A1, buttressing the finding that its expression was affected by gravity. These results suggest that a meta-analysis of DNA microarray datasets may help identify new target genes previously overlooked in individual microarray analyses.
Collapse
Affiliation(s)
- Yin Liang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mengxue Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yun Liu
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chen Wang
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
23
|
Juhl OJ, Buettmann EG, Friedman MA, DeNapoli RC, Hoppock GA, Donahue HJ. Update on the effects of microgravity on the musculoskeletal system. NPJ Microgravity 2021; 7:28. [PMID: 34301942 PMCID: PMC8302614 DOI: 10.1038/s41526-021-00158-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
With the reignited push for manned spaceflight and the development of companies focused on commercializing spaceflight, increased human ventures into space are inevitable. However, this venture would not be without risk. The lower gravitational force, known as microgravity, that would be experienced during spaceflight significantly disrupts many physiological systems. One of the most notably affected systems is the musculoskeletal system, where exposure to microgravity causes both bone and skeletal muscle loss, both of which have significant clinical implications. In this review, we focus on recent advancements in our understanding of how exposure to microgravity affects the musculoskeletal system. We will focus on the catabolic effects microgravity exposure has on both bone and skeletal muscle cells, as well as their respective progenitor stem cells. Additionally, we report on the mechanisms that underlie bone and muscle tissue loss resulting from exposure to microgravity and then discuss current countermeasures being evaluated. We reveal the gaps in the current knowledge and expound upon how current research is filling these gaps while also identifying new avenues of study as we continue to pursue manned spaceflight.
Collapse
Affiliation(s)
- Otto J Juhl
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Evan G Buettmann
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael A Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Rachel C DeNapoli
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Gabriel A Hoppock
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Henry J Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
24
|
Zhang CY, Yang CQ, Chen Q, Liu J, Zhang G, Dong C, Liu XL, Farooq HMU, Zhao SQ, Luo LH, Jiang SF, Niu YB, Yin DC. miR-194-Loaded Gelatin Nanospheres Target MEF2C to Suppress Muscle Atrophy in a Mechanical Unloading Model. Mol Pharm 2021; 18:2959-2973. [PMID: 34189919 DOI: 10.1021/acs.molpharmaceut.1c00121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Muscle atrophy usually occurs under mechanical unloading, which increases the risk of injury to reduce the functionality of the moving system, while there is still no effective therapy until now. It was found that miR-194 was significantly downregulated in a muscle atrophy model, and its target protein was the myocyte enhancer factor 2C (MEF2C). miR-194 could promote muscle differentiation and also inhibit ubiquitin ligases, thus miR-194 could be used as a nucleic acid drug to treat muscle atrophy, whereas miRNA was unstable in vivo, limiting its application as a therapeutic drug. A gelatin nanosphere (GN) delivery system was applied for the first time to load exogenous miRNA here. Exogenous miR-194 was loaded in GNs and injected into the muscle atrophy model. It demonstrated that the muscle fiber cross-sectional area, in situ muscle contractile properties, and myogenic markers were increased significantly after treatment. It proposed miR-194 loaded in GNs as an effective treatment for muscle atrophy by promoting muscle differentiation and inhibiting ubiquitin ligase activity. Moreover, the developed miRNA delivery system, taking advantage of its tunable composition, degradation rate, and capacity to load various drug molecules with high dosage, is considered a promising platform to achieve precise treatment of muscle atrophy-related diseases.
Collapse
Affiliation(s)
- Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Chang-Qing Yang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Qiang Chen
- State Key Laboratory of Solidification Processing, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Jie Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Chen Dong
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Xin-Li Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Hafiz Muhammad Umer Farooq
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Shi-Qi Zhao
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Li-Heng Luo
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Shan-Feng Jiang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Yin-Bo Niu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| |
Collapse
|
25
|
Furukawa S, Chatani M, Higashitani A, Higashibata A, Kawano F, Nikawa T, Numaga-Tomita T, Ogura T, Sato F, Sehara-Fujisawa A, Shinohara M, Shimazu T, Takahashi S, Watanabe-Takano H. Findings from recent studies by the Japan Aerospace Exploration Agency examining musculoskeletal atrophy in space and on Earth. NPJ Microgravity 2021; 7:18. [PMID: 34039989 PMCID: PMC8155041 DOI: 10.1038/s41526-021-00145-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/25/2021] [Indexed: 11/09/2022] Open
Abstract
The musculoskeletal system provides the body with correct posture, support, stability, and mobility. It is composed of the bones, muscles, cartilage, tendons, ligaments, joints, and other connective tissues. Without effective countermeasures, prolonged spaceflight under microgravity results in marked muscle and bone atrophy. The molecular and physiological mechanisms of this atrophy under unloaded conditions are gradually being revealed through spaceflight experiments conducted by the Japan Aerospace Exploration Agency using a variety of model organisms, including both aquatic and terrestrial animals, and terrestrial experiments conducted under the Living in Space project of the Japan Ministry of Education, Culture, Sports, Science, and Technology. Increasing our knowledge in this field will lead not only to an understanding of how to prevent muscle and bone atrophy in humans undergoing long-term space voyages but also to an understanding of countermeasures against age-related locomotive syndrome in the elderly.
Collapse
Affiliation(s)
- Satoshi Furukawa
- Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency, Tsukuba, Ibaraki, Japan.
| | - Masahiro Chatani
- Department of Pharmacology, Showa University School of Dentistry, Tokyo, Japan. .,Pharmacological Research Center, Showa University, Tokyo, Japan.
| | | | - Akira Higashibata
- Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency, Tsukuba, Ibaraki, Japan
| | - Fuminori Kawano
- Graduate School of Health Sciences, Matsumoto University, Matsumoto, Nagano, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Takuro Numaga-Tomita
- Department of Molecular Pharmacology, School of Medicine, Shinshu University, Matsumoto, Nagano, Japan
| | - Toshihiko Ogura
- Department of Developmental Neurobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Fuminori Sato
- Department of Growth Regulation, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Atsuko Sehara-Fujisawa
- Department of Growth Regulation, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masahiro Shinohara
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | | | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Haruko Watanabe-Takano
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
26
|
Okada R, Fujita SI, Suzuki R, Hayashi T, Tsubouchi H, Kato C, Sadaki S, Kanai M, Fuseya S, Inoue Y, Jeon H, Hamada M, Kuno A, Ishii A, Tamaoka A, Tanihata J, Ito N, Shiba D, Shirakawa M, Muratani M, Kudo T, Takahashi S. Transcriptome analysis of gravitational effects on mouse skeletal muscles under microgravity and artificial 1 g onboard environment. Sci Rep 2021; 11:9168. [PMID: 33911096 PMCID: PMC8080648 DOI: 10.1038/s41598-021-88392-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Spaceflight causes a decrease in skeletal muscle mass and strength. We set two murine experimental groups in orbit for 35 days aboard the International Space Station, under artificial earth-gravity (artificial 1 g; AG) and microgravity (μg; MG), to investigate whether artificial 1 g exposure prevents muscle atrophy at the molecular level. Our main findings indicated that AG onboard environment prevented changes under microgravity in soleus muscle not only in muscle mass and fiber type composition but also in the alteration of gene expression profiles. In particular, transcriptome analysis suggested that AG condition could prevent the alterations of some atrophy-related genes. We further screened novel candidate genes to reveal the muscle atrophy mechanism from these gene expression profiles. We suggest the potential role of Cacng1 in the atrophy of myotubes using in vitro and in vivo gene transductions. This critical project may accelerate the elucidation of muscle atrophy mechanisms.
Collapse
Affiliation(s)
- Risa Okada
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Ibaraki, 305-8505, Japan
| | - Shin-Ichiro Fujita
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Riku Suzuki
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Takuto Hayashi
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Hirona Tsubouchi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Chihiro Kato
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Master's Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Shunya Sadaki
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Maho Kanai
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Sayaka Fuseya
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Yuri Inoue
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Hyojung Jeon
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Michito Hamada
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akihiro Kuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akiko Ishii
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Jun Tanihata
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Naoki Ito
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, 650-0047, Japan
| | - Dai Shiba
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Ibaraki, 305-8505, Japan
| | - Masaki Shirakawa
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Ibaraki, 305-8505, Japan
| | - Masafumi Muratani
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Takashi Kudo
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan.
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
| | - Satoru Takahashi
- Mouse Epigenetics Project, ISS/Kibo Experiment, Japan Aerospace Exploration Agency (JAXA), Ibaraki, 305-8505, Japan.
- Laboratory Animal Resource Center in Transborder Medical Research Center, Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
27
|
Yan X, Gao X, Niu Q, Peng X, Zhang J, Ma X, Wei Y, Wang H, Gao Y, Chang H. Differential protein metabolism and regeneration in hypertrophic diaphragm and atrophic gastrocnemius muscles in hibernating Daurian ground squirrels. Exp Physiol 2021; 106:958-971. [PMID: 33517584 DOI: 10.1113/ep089187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 01/26/2021] [Indexed: 12/23/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim was to investigate whether diaphragm hypertrophy and gastrocnemius atrophy during hibernation of Daurian ground squirrels involve differential regulation of protein metabolism and regeneration. What is the main finding and its importance? We clarified the differences in protein metabolism and muscle regenerative potential in the diaphragm and gastrocnemius of hibernating ground squirrels, reflecting the different adaptability of muscles. ABSTRACT Are differences in the regulation of protein metabolism and regeneration involved in the different phenotypic adaptation mechanisms of muscle hypertrophy and atrophy in hibernators? Two fast-type muscles (diaphragm and gastrocnemius) in summer active and hibernating Daurian ground squirrels were selected to detect changes in cross-sectional area (CSA) and protein expression indicative of protein synthesis metabolism (protein expression of P-Akt, P-mTORC1, P-S6K1 and P-4E-BP1), protein degradation metabolism (MuRF1, atrogin-1, calpain-1, calpain-2, calpastatin, desmin, troponin T, Beclin1 and LC3-II) and muscle regeneration (MyoD, myogenin and myostatin). In the hibernation group compared with the summer active group, the CSA of the diaphragm muscle increased significantly by 26.1%, whereas the CSA of the gastrocnemius muscle decreased significantly by 20.4%. Our study also indicated that increased protein synthesis, decreased protein degradation and increased muscle regenerative potential contributed to diaphragm muscle hypertrophy, whereas decreased protein synthesis, increased protein degradation and decreased muscle regenerative potential contributed to gastrocnemius muscle atrophy. In conclusion, the differences in muscle regeneration and regulatory pattern of protein metabolism might contribute to the different adaptive changes observed in the diaphragm and gastrocnemius muscles of ground squirrels.
Collapse
Affiliation(s)
- Xia Yan
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Xuli Gao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Qiaohua Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Xin Peng
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Jie Zhang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Xiufeng Ma
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Yanhong Wei
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China.,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Huiping Wang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Yunfang Gao
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| | - Hui Chang
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, 710069, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (College of Life Sciences, Northwest University), Ministry of Education, Xi'an, 710069, China
| |
Collapse
|
28
|
Afshinnekoo E, Scott RT, MacKay MJ, Pariset E, Cekanaviciute E, Barker R, Gilroy S, Hassane D, Smith SM, Zwart SR, Nelman-Gonzalez M, Crucian BE, Ponomarev SA, Orlov OI, Shiba D, Muratani M, Yamamoto M, Richards SE, Vaishampayan PA, Meydan C, Foox J, Myrrhe J, Istasse E, Singh N, Venkateswaran K, Keune JA, Ray HE, Basner M, Miller J, Vitaterna MH, Taylor DM, Wallace D, Rubins K, Bailey SM, Grabham P, Costes SV, Mason CE, Beheshti A. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 2020; 183:1162-1184. [PMID: 33242416 PMCID: PMC8441988 DOI: 10.1016/j.cell.2020.10.050] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.
Collapse
Affiliation(s)
- Ebrahim Afshinnekoo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Matthew J MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eloise Pariset
- Universities Space Research Association (USRA), Mountain View, CA 94043, USA; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Richard Barker
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | - Simon Gilroy
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | | | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sara R Zwart
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mayra Nelman-Gonzalez
- KBR, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Brian E Crucian
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sergey A Ponomarev
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Oleg I Orlov
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Ibaraki 305-8505, Japan
| | - Masafumi Muratani
- Transborder Medical Research Center, and Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Stephanie E Richards
- Bionetics, NASA Kennedy Space Center, Kennedy Space Center, Merritt Island, FL 32899, USA
| | - Parag A Vaishampayan
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Myrrhe
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Eric Istasse
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Nitin Singh
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Kasthuri Venkateswaran
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Jessica A Keune
- Space Medicine Operations Division, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Hami E Ray
- ASRC Federal Space and Defense, Inc., Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Mathias Basner
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jack Miller
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Martha Hotz Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL 60208, USA; Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanne M Taylor
- Department of Biomedical Informatics, The Children's Hospital of Philadelphia, PA 19104, USA; Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen Rubins
- Astronaut Office, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Susan M Bailey
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Peter Grabham
- Center for Radiological Research, Department of Oncology, College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA; The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA.
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
29
|
Brzeszczyńska J, Brzeszczyński F, Hamilton DF, McGregor R, Simpson AHRW. Role of microRNA in muscle regeneration and diseases related to muscle dysfunction in atrophy, cachexia, osteoporosis, and osteoarthritis. Bone Joint Res 2020; 9:798-807. [PMID: 33174473 PMCID: PMC7672326 DOI: 10.1302/2046-3758.911.bjr-2020-0178.r1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that have emerged as potential predictive, prognostic, and therapeutic biomarkers, relevant to many pathophysiological conditions including limb immobilization, osteoarthritis, sarcopenia, and cachexia. Impaired musculoskeletal homeostasis leads to distinct muscle atrophies. Understanding miRNA involvement in the molecular mechanisms underpinning conditions such as muscle wasting may be critical to developing new strategies to improve patient management. MicroRNAs are powerful post-transcriptional regulators of gene expression in muscle and, importantly, are also detectable in the circulation. MicroRNAs are established modulators of muscle satellite stem cell activation, proliferation, and differentiation, however, there have been limited human studies that investigate miRNAs in muscle wasting. This narrative review summarizes the current knowledge as to the role of miRNAs in the skeletal muscle differentiation and atrophy, synthesizing the findings of published data. Cite this article: Bone Joint Res 2020;9(11):798-807.
Collapse
Affiliation(s)
- Joanna Brzeszczyńska
- School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
- Department of Molecular Biophysics, University of Lodz, Lodz, Poland
| | | | - David F Hamilton
- School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Robin McGregor
- Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, South Korea
| | | |
Collapse
|
30
|
MicroRNA molecules as predictive biomarkers of adaptive responses to strength training and physical inactivity in haemodialysis patients. Sci Rep 2020; 10:15597. [PMID: 32973233 PMCID: PMC7519115 DOI: 10.1038/s41598-020-72542-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/24/2020] [Indexed: 01/31/2023] Open
Abstract
The miRNA-206 and miRNA-23a play an important role in muscle tissue hypertrophy, regeneration and atrophy. Both of these miRNAs have been highlighted as promising adaptation predictors; however, the available evidence on associations is inconclusive. Therefore, our aim was to assess the expression levels of these two miRNAs as predictors of change in muscle function during strength training and physical inactivity among dialysed patients. For this purpose, 46 haemodialysis patients were monitored for 12-weeks of either intradialytic strength training (EXG, n = 20) or physical inactivity during dialysis (CON, n = 26). In both groups of patients, we assessed the baseline expression levels of miRNA-23a and miRNA-206 and the isometric force generated during hip flexion (HF) contraction before and after the 12-week period. Among the EXG group, the expression of miRNA-206 predicted the change in HF (R2 = 0.63, p = 0.0005) much more strongly than the expression of miRNA-23a (R2 = 0.21, p = 0.027). Interestingly, baseline miRNA-23a (R2 = 0.30, p = 0.006) predicted the change in HF much more than miRNA-206 (p = ns) among the CON group. Our study indicates that the baseline expression of miRNA-206 could predict the response to strength training, while miRNA-23a could serve as a potential predictive marker of functional changes during physical inactivity in dialysis patients.
Collapse
|
31
|
Bjorkman KK, Guess MG, Harrison BC, Polmear MM, Peter AK, Leinwand LA. miR-206 enforces a slow muscle phenotype. J Cell Sci 2020; 133:jcs243162. [PMID: 32620696 PMCID: PMC7438006 DOI: 10.1242/jcs.243162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/25/2020] [Indexed: 12/21/2022] Open
Abstract
Striated muscle is a highly specialized collection of tissues with contractile properties that vary according to functional needs. Although muscle fiber types are established postnatally, lifelong plasticity facilitates stimulus-dependent adaptation. Functional adaptation requires molecular adaptation, which is partially provided by miRNA-mediated post-transcriptional regulation. miR-206 is a muscle-specific miRNA enriched in slow muscles. We investigated whether miR-206 drives the slow muscle phenotype or is merely an outcome. We found that miR-206 expression increases in both physiological (including female sex and endurance exercise) and pathological conditions (muscular dystrophy and adrenergic agonism) that promote a slow phenotype. Consistent with that observation, the slow soleus muscle of male miR-206-knockout mice displays a faster phenotype than wild-type mice. Moreover, left ventricles of male miR-206 knockout mice have a faster myosin profile, accompanied by dilation and systolic dysfunction. Thus, miR-206 appears to be necessary to enforce a slow skeletal and cardiac muscle phenotype and to play a key role in muscle sexual dimorphisms.
Collapse
Affiliation(s)
- Kristen K Bjorkman
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Martin G Guess
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Brooke C Harrison
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Michael M Polmear
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Angela K Peter
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular, and Developmental Biology, BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave., UCB596, Boulder, CO 80303, USA
| |
Collapse
|
32
|
Grimm D, Wehland M, Corydon TJ, Richter P, Prasad B, Bauer J, Egli M, Kopp S, Lebert M, Krüger M. The effects of microgravity on differentiation and cell growth in stem cells and cancer stem cells. Stem Cells Transl Med 2020; 9:882-894. [PMID: 32352658 PMCID: PMC7381804 DOI: 10.1002/sctm.20-0084] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/12/2022] Open
Abstract
A spaceflight has enormous influence on the health of space voyagers due to the combined effects of microgravity and cosmic radiation. Known effects of microgravity (μg) on cells are changes in differentiation and growth. Considering the commercialization of spaceflight, future space exploration, and long-term manned flights, research focusing on differentiation and growth of stem cells and cancer cells exposed to real (r-) and simulated (s-) μg is of high interest for regenerative medicine and cancer research. In this review, we focus on platforms to study r- and s-μg as well as the impact of μg on cancer stem cells in the field of gastrointestinal cancer, lung cancer, and osteosarcoma. Moreover, we review the current knowledge of different types of stem cells exposed to μg conditions with regard to differentiation and engineering of cartilage, bone, vasculature, heart, skin, and liver constructs.
Collapse
Affiliation(s)
- Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, Magdeburg, Germany.,Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Markus Wehland
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany
| | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Richter
- Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Binod Prasad
- Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johann Bauer
- Max Planck Institute of Biochemistry, Planegg-Martinsried, Germany
| | - Marcel Egli
- Institute of Medical Engineering, Space Biology Group, Lucerne University of Applied Sciences and Arts, Hergiswil, Switzerland
| | - Sascha Kopp
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany
| | - Michael Lebert
- Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany.,Space Biology Unlimited SAS, Bordeaux, France
| | - Marcus Krüger
- Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
33
|
Validation of a New Rodent Experimental System to Investigate Consequences of Long Duration Space Habitation. Sci Rep 2020; 10:2336. [PMID: 32047211 PMCID: PMC7012842 DOI: 10.1038/s41598-020-58898-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Animal models are useful for exploring the health consequences of prolonged spaceflight. Capabilities were developed to perform experiments in low earth orbit with on-board sample recovery, thereby avoiding complications caused by return to Earth. For NASA’s Rodent Research-1 mission, female mice (ten 32 wk C57BL/6NTac; ten 16 wk C57BL/6J) were launched on an unmanned vehicle, then resided on the International Space Station for 21/22d or 37d in microgravity. Mice were euthanized on-orbit, livers and spleens dissected, and remaining tissues frozen in situ for later analyses. Mice appeared healthy by daily video health checks and body, adrenal, and spleen weights of 37d-flight (FLT) mice did not differ from ground controls housed in flight hardware (GC), while thymus weights were 35% greater in FLT than GC. Mice exposed to 37d of spaceflight displayed elevated liver mass (33%) and select enzyme activities compared to GC, whereas 21/22d-FLT mice did not. FLT mice appeared more physically active than respective GC while soleus muscle showed expected atrophy. RNA and enzyme activity levels in tissues recovered on-orbit were of acceptable quality. Thus, this system establishes a new capability for conducting long-duration experiments in space, enables sample recovery on-orbit, and avoids triggering standard indices of chronic stress.
Collapse
|
34
|
McDonald JT, Stainforth R, Miller J, Cahill T, da Silveira WA, Rathi KS, Hardiman G, Taylor D, Costes SV, Chauhan V, Meller R, Beheshti A. NASA GeneLab Platform Utilized for Biological Response to Space Radiation in Animal Models. Cancers (Basel) 2020; 12:E381. [PMID: 32045996 PMCID: PMC7072278 DOI: 10.3390/cancers12020381] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Ionizing radiation from galactic cosmic rays (GCR) is one of the major risk factors that will impact the health of astronauts on extended missions outside the protective effects of the Earth's magnetic field. The NASA GeneLab project has detailed information on radiation exposure using animal models with curated dosimetry information for spaceflight experiments. Methods: We analyzed multiple GeneLab omics datasets associated with both ground-based and spaceflight radiation studies that included in vivo and in vitro approaches. A range of ions from protons to iron particles with doses from 0.1 to 1.0 Gy for ground studies, as well as samples flown in low Earth orbit (LEO) with total doses of 1.0 mGy to 30 mGy, were utilized. Results: From this analysis, we were able to identify distinct biological signatures associating specific ions with specific biological responses due to radiation exposure in space. For example, we discovered changes in mitochondrial function, ribosomal assembly, and immune pathways as a function of dose. Conclusions: We provided a summary of how the GeneLab's rich database of omics experiments with animal models can be used to generate novel hypotheses to better understand human health risks from GCR exposures.
Collapse
Affiliation(s)
| | - Robert Stainforth
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Jack Miller
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| | - Thomas Cahill
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Willian A. da Silveira
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
| | - Komal S. Rathi
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Gary Hardiman
- School of Biological Sciences & Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK; (T.C.); (W.A.d.S.)
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Deanne Taylor
- Department of Biomedical Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvain V. Costes
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA 94035, USA;
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, ON K1A-1C1, Canada; (R.S.); (V.C.)
| | - Robert Meller
- Department of Neurobiology and Pharmacology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Afshin Beheshti
- KBR, NASA Ames Research Center, Moffett Field, CA 94035, USA;
| |
Collapse
|
35
|
Bao T, Han H, Li B, Zhao Y, Bou G, Zhang X, Du M, Zhao R, Mongke T, Laxima, Ding W, Jia Z, Dugarjaviin M, Bai D. The distinct transcriptomes of fast-twitch and slow-twitch muscles in Mongolian horses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 33:100649. [PMID: 31869634 DOI: 10.1016/j.cbd.2019.100649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/19/2023]
Abstract
Skeletal muscle is the largest organ system in the mammalian body and plays a key role in locomotion of horses. Fast and slow muscle fibers have different abilities and functions to adapt to exercises. To investigate the RNA and miRNA expression profiles in the muscles with different muscle fiber compositions on Mongolian horses. We examined the muscle fiber type population and produced deep RNA sequencing for different parts of skeletal muscles. And chose two of them with the highest difference in fast and slow muscle fiber population (splenius and gluteus medius) for comparing the gene expression profile of slow and fast muscle fiber types. We identified a total of 275 differentially expressed genes (DEGs), and 11 differentially expressed miRNAs (DEmiRs). In addition, target gene prediction and alternative splicing analysis were also performed. Significant correlations were found between the differentially expressed gene, miRNAs, and alternative splicing events. The result indicated that differentially expressed muscle-specific genes and target genes of miRNAs might co-regulating the performance of slow and fast muscle fiber types in Mongolian horses.
Collapse
Affiliation(s)
- Tugeqin Bao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Haige Han
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Bei Li
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yiping Zhao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Gerelchimeg Bou
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xinzhuang Zhang
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ming Du
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ruoyang Zhao
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Togtokh Mongke
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Laxima
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wenqi Ding
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Zijie Jia
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Manglai Dugarjaviin
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Dongyi Bai
- Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Hohhot 010018, China; Scientific Observing and Experimental Station of Equine Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China; Equine Research Center, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China.
| |
Collapse
|
36
|
Baek MO, Ahn CB, Cho HJ, Choi JY, Son KH, Yoon MS. Simulated microgravity inhibits C2C12 myogenesis via phospholipase D2-induced Akt/FOXO1 regulation. Sci Rep 2019; 9:14910. [PMID: 31624287 PMCID: PMC6797799 DOI: 10.1038/s41598-019-51410-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/30/2019] [Indexed: 12/19/2022] Open
Abstract
The skeletal muscle system has evolved to maintain body posture against a constant gravitational load. Mammalian target of rapamycin (mTOR) regulates the mechanically induced increase in the skeletal muscle mass. In the present study, we investigated mTOR pathway in C2C12 myoblasts in a model of mechanical unloading by creating a simulated microgravity (SM) using 3 D clinorotation. SM decreased the phosphorylation of Akt at Ser 473, which was mediated by mTOR complex 2 (mTORC2), in C2C12 myoblasts, leading to a decrease in the cell growth rate. Subsequently, SM inhibited C2C12 myogenesis in an Akt-dependent manner. In addition, SM increased the phospholipase D (PLD) activity by enhancing PLD2 expression, resulting in the dissociation of mSIN1 from the mTORC2, followed by decrease in the phosphorylation of Akt at Ser 473, and FOXO1 at Ser 256 in C2C12 myoblasts. Exposure to SM decreased the autophagic flux of C2C12 myoblasts by regulation of mRNA level of autophagic genes in a PLD2 and FOXO1-dependent manner, subsequently, resulting in a decrease in the C2C12 myogenesis. In conclusion, by analyzing the molecular signature of C2C12 myogenesis using SM, we suggest that the regulatory axis of the PLD2 induced Akt/FOXO1, is critical for C2C12 myogenesis.
Collapse
Affiliation(s)
- Mi-Ock Baek
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Chi Bum Ahn
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Hye-Jeong Cho
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Ji-Young Choi
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon, 21565, Republic of Korea.
| | - Mee-Sup Yoon
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea. .,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, Republic of Korea. .,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 21999, Republic of Korea.
| |
Collapse
|
37
|
An L, Li Y, Fan Y, He N, Ran F, Qu H, Wang Y, Zhao X, Ye C, Jiang Y, Fang X, Hang H. The Trends in Global Gene Expression in Mouse Embryonic Stem Cells During Spaceflight. Front Genet 2019; 10:768. [PMID: 31552089 PMCID: PMC6743352 DOI: 10.3389/fgene.2019.00768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The environment in space differs greatly from the environment on the ground. Spaceflight causes a number of physiological changes in astronauts, such as bone loss and immune system dysregulation. These effects threaten astronauts’ space missions, and understanding the underlying cellular and molecular mechanisms is important to manage the risks of space missions. The biological effects of spaceflight on mammalian cells, especially with regards to DNA damage, have attracted much attention. Rad9−/− mouse embryonic stem cells (mESCs) are known to be extremely sensitive to DNA damage agents. In this study, a project of the SJ-10 satellite programme, we investigated the gene expression profiles of both Rad9−/− mESCs and Rad9+/+ (wild-type) mESCs in space with a focus on genes critical for inducing, preventing, or repairing genomic DNA lesions. We found that spaceflight downregulated more genes than it upregulated in both wild-type and Rad9−/− mESCs, indicating a suppressive effect of spaceflight on global gene expression. In contrast, Rad9 deletion upregulated more genes than it downregulated. Of note, spaceflight mainly affected organ development and influenced a wide range of cellular functions in mESCs, while Rad9 deletion mainly affected the development and function of the hematological system, especially the development, differentiation and function of immune cells. The patterns of gene expression in mouse embryonic stem cells in space is distinct from those in other types of cells. In addition, both spaceflight and Rad9 deletion downregulated DNA repair genes, suggesting a possibility that spaceflight has negative effects on genome for embryonic stem cells and the effects are likely worsen when the genome maintenance mechanism is defective.
Collapse
Affiliation(s)
- Lili An
- Key Laboratory of Protein and Peptide Drugs, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yanming Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yingjun Fan
- Key Laboratory of Protein and Peptide Drugs, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ning He
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Fanlei Ran
- Key Laboratory of Protein and Peptide Drugs, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hongzhu Qu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yanqiu Wang
- Center for Space Science and Applied Research, Chinese Academy of Sciences, Beijing, China
| | - Xuetong Zhao
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chen Ye
- Key Laboratory of Protein and Peptide Drugs, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yuanda Jiang
- Center for Space Science and Applied Research, Chinese Academy of Sciences, Beijing, China
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Haiying Hang
- Key Laboratory of Protein and Peptide Drugs, National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
38
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
39
|
Fu S, Meng Y, Zhang W, Wang J, He Y, Huang L, Chen H, Kuang J, Du H. Transcriptomic Responses of Skeletal Muscle to Acute Exercise in Diabetic Goto-Kakizaki Rats. Front Physiol 2019; 10:872. [PMID: 31338039 PMCID: PMC6629899 DOI: 10.3389/fphys.2019.00872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/21/2019] [Indexed: 12/27/2022] Open
Abstract
Physical activity exerts positive effects on glycemic control in type 2 diabetes (T2D), which is mediated in part by extensive metabolic and molecular remodeling of skeletal muscle in response to exercise, while many regulators of skeletal muscle remain unclear. In the present study, we investigated the effects of acute exercise on skeletal muscle transcriptomic responses in the Goto-Kakizaki (GK) rats which can spontaneously develop T2D. The transcriptomes of skeletal muscle from both 8-week-old GK and Wistar rats that underwent a single exercise session (60 min running using an animal treadmill at 15 m/min) or remained sedentary were analyzed by next-generation RNA sequencing. We identified 819 differentially expressed genes in the sedentary GK rats compared with those of the sedentary Wistar rats. After a single bout of running, we found 291 and 598 genes that were differentially expressed in the exercise GK and exercise Wistar rats when compared with the corresponding sedentary rats. By integrating our data and previous studies including RNA or protein expression patterns and transgenic experiments, the downregulated expression of Fasn and upregulated expression of Tbc1d1, Hk2, Lpin1, Ppargc1a, Sorbs1, and Hmox1 might enhance glucose uptake or improve insulin sensitivity to ameliorate hyperglycemia in the exercise GK rats. Our results provide mechanistic insight into the beneficial effects of exercise on hyperglycemia and insulin action in skeletal muscle of diabetic GK rats.
Collapse
Affiliation(s)
- Shuying Fu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yuhuan Meng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Wenlu Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Jiajian Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yuting He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongmei Chen
- Department of Endocrinology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian Kuang
- Department of Endocrinology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
40
|
Skeletal muscle in MuRF1 null mice is not spared in low-gravity conditions, indicating atrophy proceeds by unique mechanisms in space. Sci Rep 2019; 9:9397. [PMID: 31253821 PMCID: PMC6599046 DOI: 10.1038/s41598-019-45821-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
Microgravity exposure is associated with loss of muscle mass and strength. The E3 ubiquitin ligase MuRF1 plays an integral role in degrading the contractile apparatus of skeletal muscle; MuRF1 null (KO) mice have shown protection in ground-based models of muscle atrophy. In contrast, MuRF1 KO mice subjected to 21 days of microgravity on the International Space Station (ISS) were not protected from muscle atrophy. In a time course experiment microgravity-induced muscle loss on the ISS showed MuRF1 gene expression was not upregulated. A comparison of the soleus transcriptome profiles between spaceflight and a publicly available data set for hindlimb suspension, a claimed surrogate model of microgravity, showed only marginal commonalities between the models. These findings demonstrate spaceflight induced atrophy is unique, and that understanding of effects of space requires study situated beyond the Earth’s mesosphere.
Collapse
|
41
|
Favia M, Fitak R, Guerra L, Pierri CL, Faye B, Oulmouden A, Burger PA, Ciani E. Beyond the Big Five: Investigating Myostatin Structure, Polymorphism and Expression in Camelus dromedarius. Front Genet 2019; 10:502. [PMID: 31231423 PMCID: PMC6566074 DOI: 10.3389/fgene.2019.00502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Myostatin, a negative regulator of skeletal muscle mass in animals, has been shown to play a role in determining muscular hypertrophy in several livestock species, and a high degree of polymorphism has been previously reported for this gene in humans and cattle. In this study, we provide a characterization of the myostatin gene in the dromedary (Camelus dromedarius) at the genomic, transcript and protein level. The gene was found to share high structural and sequence similarity with other mammals, notably Old World camelids. 3D modeling highlighted several non-conservative SNP variants compared to the bovine, as well as putative functional variants involved in the stability of the myostatin dimer. NGS data for nine dromedaries from various countries revealed 66 novel SNPs, all of them falling either upstream or downstream the coding region. The analysis also confirmed the presence of three previously described SNPs in intron 1, predicted here to alter both splicing and transcription factor binding sites (TFBS), thus possibly impacting myostatin processing and/or regulation. Several putative TFBS were identified in the myostatin upstream region, some of them belonging to the myogenic regulatory factor family. Patterns of SNP distribution across countries, as suggested by Bayesian clustering of the nine dromedaries using the 69 SNPs, pointed to weak geographic differentiation, in line with known recurrent gene flow at ancient trading centers along caravan routes. Myostatin expression was investigated in a set of 8 skeletal muscles, both at transcript and protein level, via Digital Droplet PCR and Western Blotting, respectively. No significant differences were observed at the transcript level, while, at the protein level, the only significant differences concerned the promyostatin dimer (75 kDa), in four pair-wise comparisons, all involving the tensor fasciae latae muscle. Beside the mentioned band at 75 kDa, additional bands were observed at around 40 and 25 kDa, corresponding to the promyostatin monomer and the active C-terminal myostatin dimer, respectively. Their weaker intensity suggests that the unprocessed myostatin dimers could act as important reservoirs of slowly available myostatin forms. Under this assumption, the sequential cleavage steps may contribute additional layers of control within an already complex regulatory framework.
Collapse
Affiliation(s)
- Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Robert Fitak
- Research Institute of Wildlife Ecology, Vetmeduni, Vienna, Austria.,Department of Biology, Duke University, Durham, NC, United States
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | | | - Ahmad Oulmouden
- Département Sciences du Vivant, Université de Limoges, Limoges, France
| | | | - Elena Ciani
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
42
|
Muscle Atrophy Marker Expression Differs between Rotary Cell Culture System and Animal Studies. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2042808. [PMID: 30906768 PMCID: PMC6398068 DOI: 10.1155/2019/2042808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/06/2018] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
Muscular atrophy, defined as the loss of muscle tissue, is a serious issue for immobilized patients on Earth and for humans during spaceflight, where microgravity prevents normal muscle loading. In vitro modeling is an important step in understanding atrophy mechanisms and testing countermeasures before animal trials. The most ideal environment for modeling must be empirically determined to best mimic known responses in vivo. To simulate microgravity conditions, murine C2C12 myoblasts were cultured in a rotary cell culture system (RCCS). Alginate encapsulation was compared against polystyrene microcarrier beads as a substrate for culturing these adherent muscle cells. Changes after culture under simulated microgravity were characterized by assessing mRNA expression of MuRF1, MAFbx, Caspase 3, Akt2, mTOR, Ankrd1, and Foxo3. Protein concentration of myosin heavy chain 4 (Myh4) was used as a differentiation marker. Cell morphology and substrate structure were evaluated with brightfield and fluorescent imaging. Differentiated C2C12 cells encapsulated in alginate had a significant increase in MuRF1 only following simulated microgravity culture and were morphologically dissimilar to normal cultured muscle tissue. On the other hand, C2C12 cells cultured on polystyrene microcarriers had significantly increased expression of MuRF1, Caspase 3, and Foxo3 and easily identifiable multinucleated myotubes. The extent of differentiation was higher in simulated microgravity and protein synthesis more active with increased Myh4, Akt2, and mTOR. The in vitro microcarrier model described herein significantly increases expression of several of the same atrophy markers as in vivo models. However, unlike animal models, MAFbx and Ankrd1 were not significantly increased and the fold change in MuRF1 and Foxo3 was lower than expected. Using a standard commercially available RCCS, the substrates and culture methods described only partially model changes in mRNAs associated with atrophy in vivo.
Collapse
|
43
|
Teodori L, Costa A, Campanella L, Albertini MC. Skeletal Muscle Atrophy in Simulated Microgravity Might Be Triggered by Immune-Related microRNAs. Front Physiol 2019; 9:1926. [PMID: 30687129 PMCID: PMC6335973 DOI: 10.3389/fphys.2018.01926] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/21/2018] [Indexed: 02/01/2023] Open
Abstract
Exposure to microgravity induces skeletal muscle disorders including atrophy, muscle force decrease, fiber-type shift. Microgravity also contributes to immune-function alterations and modifies microRNAs (miRs) expression. To understand the link between microgravity-induced skeletal muscle atrophy and immune function deregulation, a bioinformatics study was performed. The web platform MiRNet was used for miRs-targets interaction analysis from previous proteomic studies on human soleus (SOL) and vastus lateralis (VL) muscles. We predicted miRs targeting deregulated gene expression following bed rest as a model of microgravity exposure; namely, let-7a-5p, miR-125b-5p for over-expressed genes in SOL and VL; miR-1-3p, miR-125b-5p and miR-1-3p, miR-95-5p for down-expressed genes in VL and SOL. The predicted miRs have important immune functions, exhibiting a significant role on both inflammation and atrophy. Let-7a down-expression leads to proliferation pathways promotion and differentiation pathway inhibition, whereas miR-1-3p over-expression yields anti-proliferative effect, promoting early differentiation. Such conflicting signals could lead to impairment between proliferation and differentiation in skeletal muscles. Moreover, promotion of an M2-like macrophage phenotype (IL-13, IL-10) by let-7a down-regulation and simultaneous promotion of an M1-like macrophage (IL-6, TNF-α) phenotype through the over-expression of EEF2 lead to a deregulation between M1/M2 tuning, that is responsible for a first pro-inflammatory/proliferative phase followed by an anti-inflammatory pro-myogenic phase during skeletal muscle regeneration after injury. These observations are important to understand the mechanism by which inflammation may play a significant role in skeletal muscle dysfunction in spaceflights, providing new links between immune response and skeletal muscle deregulation, which may be useful to further investigate possible therapeutic intervention.
Collapse
Affiliation(s)
- Laura Teodori
- Diagnostic and Metrology Laboratory, TECFIS-FSN, ENEA, Frascati, Italy
| | - Alessandra Costa
- Diagnostic and Metrology Laboratory, TECFIS-FSN, ENEA, Frascati, Italy
| | - Luigi Campanella
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Maria C Albertini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
44
|
Genchi GG, Degl'Innocenti A, Salgarella AR, Pezzini I, Marino A, Menciassi A, Piccirillo S, Balsamo M, Ciofani G. Modulation of gene expression in rat muscle cells following treatment with nanoceria in different gravity regimes. Nanomedicine (Lond) 2018; 13:2821-2833. [PMID: 30334476 DOI: 10.2217/nnm-2018-0316] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM Oxidative stress (OS) is strictly associated with senescence/pathogenesis of biological systems. As putative countermeasure to environmental OS, cerium oxide nanoparticles (nanoceria [NC]) were administered to muscle cells on ground and aboard the International Space Station. MATERIALS & METHODS Transcriptional analyses were conducted through microarray technology and hierarchical clustering. Venn diagram and gene ontology analyses were also performed on selected gene lists. RESULTS Adaptive responses to both NC administration and to permanence in real microgravity conditions occurred. Enrichment in the biological processes related to aging, body fat development and mesodermal tissue proliferation for NC-treated samples were found. CONCLUSION Nanotechnology antioxidants promise applications to pathological conditions governed by OS on Earth and in life-hostile environments (low Earth orbit and deep space).
Collapse
Affiliation(s)
- Giada Graziana Genchi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Andrea Degl'Innocenti
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Alice Rita Salgarella
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Ilaria Pezzini
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Arianna Menciassi
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy
| | - Sara Piccirillo
- Agenzia Spaziale Italiana, Via del Politecnico snc, Roma 00133, Italy
| | - Michele Balsamo
- Kayser Italia S.r.l., Via di Popogna 501, Livorno 57128, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera (Pisa) 56025, Italy.,Politecnico di Torino, Department of Aerospace & Mechanical Engineering, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| |
Collapse
|
45
|
Ray S, Gebre S, Fogle H, Berrios DC, Tran PB, Galazka JM, Costes SV. GeneLab: Omics database for spaceflight experiments. Bioinformatics 2018; 35:1753-1759. [DOI: 10.1093/bioinformatics/bty884] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Shayoni Ray
- Space Biosciences Division, USRA/NASA Ames Research Center, Moffett Field, CA, USA
| | - Samrawit Gebre
- Space Biosciences Division, KBRwyle/NASA Ames Research Center, Moffett Field, CA, USA
| | - Homer Fogle
- Space Biosciences Division, KBRwyle/NASA Ames Research Center, Moffett Field, CA, USA
| | - Daniel C Berrios
- Space Biosciences Division, USRA/NASA Ames Research Center, Moffett Field, CA, USA
| | - Peter B Tran
- Intelligent Systems Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| |
Collapse
|
46
|
A microRNA signature and TGF-β1 response were identified as the key master regulators for spaceflight response. PLoS One 2018; 13:e0199621. [PMID: 30044882 PMCID: PMC6059388 DOI: 10.1371/journal.pone.0199621] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/03/2018] [Indexed: 12/30/2022] Open
Abstract
Translating fundamental biological discoveries from NASA Space Biology program into health risk from space flights has been an ongoing challenge. We propose to use NASA GeneLab database to gain new knowledge on potential systemic responses to space. Unbiased systems biology analysis of transcriptomic data from seven different rodent datasets reveals for the first time the existence of potential “master regulators” coordinating a systemic response to microgravity and/or space radiation with TGF-β1 being the most common regulator. We hypothesized the space environment leads to the release of biomolecules circulating inside the blood stream. Through datamining we identified 13 candidate microRNAs (miRNA) which are common in all studies and directly interact with TGF-β1 that can be potential circulating factors impacting space biology. This study exemplifies the utility of the GeneLab data repository to aid in the process of performing novel hypothesis–based research.
Collapse
|
47
|
Han B, Wei SP, Zhang XC, Li H, Li Y, Li RX, Li K, Zhang XZ. Effects of constrained dynamic loading, CKIP‑1 gene knockout and combination stimulations on bone loss caused by mechanical unloading. Mol Med Rep 2018; 18:2506-2514. [PMID: 29956799 DOI: 10.3892/mmr.2018.9222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/20/2018] [Indexed: 11/05/2022] Open
Abstract
Mechanical stimulation plays an important role in maintaining the growth and normal function of the skeletal system. Mechanical unloading occurs, for example, in astronauts spending long periods of time in space or in patients on prolonged bed rest, and causes a rapid loss of bone mass. Casein kinase 2‑interacting protein‑1 (CKIP‑1) is a novel negative bone regulation factor that has been demonstrated to reduce bone loss and enhance bone formation. The aim of this study was to investigate the effect of constrained dynamic loading (Loading) in combination with CKIP‑1 gene knockout (KO) on unloading‑induced bone loss in tail‑suspension mice. The blood serum metabolism index [alkaline phosphatase (ALP) activity and osteocalcin (OCN) levels], tibia mechanical behavior (including bone trabecular microstructure parameters and tibia biomechanical properties), osteoblast‑related gene expression [ALP, OCN, collagen I and bone morphogenetic protein‑2 and osteoprotegerin (OPG)] and osteoclast‑related gene expression [receptor activators of NF‑kB ligand (RANKL)] were measured. The results demonstrated that mice experienced a loss of bone mass after four weeks of tail suspension compared with a wild type group. The mechanical properties, microarchitecture and mRNA expression were significantly increased in mice after Loading + KO treatment (P<0.05). Furthermore, compared with loading or KO alone, the ratio of OPG/RANKL was increased in the combined treatment group. The combined effect of Loading + KO was greater than that observed with loading or KO alone (P<0.05). The present study demonstrates that Loading + KO can counter unloading‑induced bone loss, and combining the two treatments has an additive effect. These results indicate that combined therapy could be a novel strategy for the clinical treatment of disuse osteoporosis associated with space travel or bed rest.
Collapse
Affiliation(s)
- Biao Han
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| | - Shu-Ping Wei
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| | - Xin-Chang Zhang
- Department of Clinical Medicine, Logistical College of People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Hao Li
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| | - Yu Li
- Department of Clinical Medicine, Logistical College of People's Armed Police Forces, Tianjin 300162, P.R. China
| | - Rui-Xin Li
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| | - Kairen Li
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| | - Xi-Zheng Zhang
- Department of Biomedical Engineering and Medical Technology, Tianjin Institute of Medical Equipment, Academy of Military Medical Sciences, Tianjin 300161, P.R. China
| |
Collapse
|
48
|
Gao Y, Arfat Y, Wang H, Goswami N. Muscle Atrophy Induced by Mechanical Unloading: Mechanisms and Potential Countermeasures. Front Physiol 2018; 9:235. [PMID: 29615929 PMCID: PMC5869217 DOI: 10.3389/fphys.2018.00235] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/02/2018] [Indexed: 12/23/2022] Open
Abstract
Prolonged periods of skeletal muscle inactivity or mechanical unloading (bed rest, hindlimb unloading, immobilization, spaceflight and reduced step) can result in a significant loss of musculoskeletal mass, size and strength which ultimately lead to muscle atrophy. With advancement in understanding of the molecular and cellular mechanisms involved in disuse skeletal muscle atrophy, several different signaling pathways have been studied to understand their regulatory role in this process. However, substantial gaps exist in our understanding of the regulatory mechanisms involved, as well as their functional significance. This review aims to update the current state of knowledge and the underlying cellular mechanisms related to skeletal muscle loss during a variety of unloading conditions, both in humans and animals. Recent advancements in understanding of cellular and molecular mechanisms, including IGF1-Akt-mTOR, MuRF1/MAFbx, FOXO, and potential triggers of disuse atrophy, such as calcium overload and ROS overproduction, as well as their role in skeletal muscle protein adaptation to disuse is emphasized. We have also elaborated potential therapeutic countermeasures that have shown promising results in preventing and restoring disuse-induced muscle loss. Finally, identified are the key challenges in this field as well as some future prospectives.
Collapse
Affiliation(s)
- Yunfang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Yasir Arfat
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Huiping Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Ministry of Education, Northwest University, Xi'an, China
| | - Nandu Goswami
- Physiology Unit, Otto Loewi Center of Research for Vascular Biology, Immunity and Inflammation, Medical University of Graz, Graz, Austria
| |
Collapse
|
49
|
Dickinson JM, D'Lugos AC, Naymik MA, Siniard AL, Wolfe AJ, Curtis DR, Huentelman MJ, Carroll CC. Transcriptome response of human skeletal muscle to divergent exercise stimuli. J Appl Physiol (1985) 2018. [PMID: 29543133 DOI: 10.1152/japplphysiol.00014.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aerobic (AE) and resistance exercise (RE) elicit unique adaptations in skeletal muscle that have distinct implications for health and performance. The purpose of this study was to identify the unique transcriptome response of skeletal muscle to acute AE and RE. In a counterbalanced, crossover design, six healthy, recreationally active young men (27 ± 3 yr) completed acute AE (40 min of cycling, ∼70% maximal HR) and RE [8 sets, 10 reps, ∼65% 1-repetition maximum (1RM)], separated by ∼1 wk. Muscle biopsies (vastus lateralis) were obtained before and at 1 and 4 h postexercise. Whole transcriptome RNA sequencing (HiSeq2500; Illumina) was performed on cDNA synthesized from skeletal muscle RNA. Sequencing data were analyzed using HTSeq, and differential gene expression was identified using DESeq2 [adjusted P value (FDR) <0.05, >1.5-fold change from preexercise]. RE resulted in a greater number of differentially expressed genes at 1 (67 vs. 48) and 4 h (523 vs. 221) compared with AE. We identified 348 genes that were differentially expressed only following RE, whereas 48 genes were differentially expressed only following AE. Gene clustering indicated that AE targeted functions related to zinc interaction, angiogenesis, and ubiquitination, whereas RE targeted functions related to transcription regulation, cytokine activity, cell adhesion, kinase activity, and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. ESRRG and TNFSRF12A were identified as potential targets related to the specific response of skeletal muscle to AE and RE, respectively. These data describe the early postexercise transcriptome response of skeletal muscle to acute AE and RE and further highlight that different forms of exercise stimulate unique molecular activity in skeletal muscle. NEW & NOTEWORTHY Whole transcriptome RNA sequencing was used to determine the early postexercise transcriptome response of skeletal muscle to acute aerobic (AE) and resistance exercise (RE) in untrained individuals. Although a number of shared genes were stimulated following both AE and RE, several genes were uniquely responsive to each exercise mode. These findings support the need for future research focused to better identify the role of exercise mode as it relates to targeting specific cellular skeletal muscle abnormalities.
Collapse
Affiliation(s)
- Jared M Dickinson
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Andrew C D'Lugos
- School of Nutrition and Health Promotion, Healthy Lifestyles Research Center, Exercise Science and Health Promotion, Arizona State University , Phoenix, Arizona
| | - Marcus A Naymik
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | - Amanda J Wolfe
- Translational Genomics Research Institute , Phoenix, Arizona
| | | | | | - Chad C Carroll
- Midwestern University , Glendale, Arizona.,Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| |
Collapse
|
50
|
Global transcriptomic analysis suggests carbon dioxide as an environmental stressor in spaceflight: A systems biology GeneLab case study. Sci Rep 2018. [PMID: 29520055 PMCID: PMC5843582 DOI: 10.1038/s41598-018-22613-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Spaceflight introduces a combination of environmental stressors, including microgravity, ionizing radiation, changes in diet and altered atmospheric gas composition. In order to understand the impact of each environmental component on astronauts it is important to investigate potential influences in isolation. Rodent spaceflight experiments involve both standard vivarium cages and animal enclosure modules (AEMs), which are cages used to house rodents in spaceflight. Ground control AEMs are engineered to match the spaceflight environment. There are limited studies examining the biological response invariably due to the configuration of AEM and vivarium housing. To investigate the innate global transcriptomic patterns of rodents housed in spaceflight-matched AEM compared to standard vivarium cages we utilized publicly available data from the NASA GeneLab repository. Using a systems biology approach, we observed that AEM housing was associated with significant transcriptomic differences, including reduced metabolism, altered immune responses, and activation of possible tumorigenic pathways. Although we did not perform any functional studies, our findings revealed a mild hypoxic phenotype in AEM, possibly due to atmospheric carbon dioxide that was increased to match conditions in spaceflight. Our investigation illustrates the process of generating new hypotheses and informing future experimental research by repurposing multiple space-flown datasets.
Collapse
|