1
|
Vinken M, Grimm D, Baatout S, Baselet B, Beheshti A, Braun M, Carstens AC, Casaletto JA, Cools B, Costes SV, De Meulemeester P, Doruk B, Eyal S, Ferreira MJS, Miranda S, Hahn C, Helvacıoğlu Akyüz S, Herbert S, Krepkiy D, Lichterfeld Y, Liemersdorf C, Krüger M, Marchal S, Ritz J, Schmakeit T, Stenuit H, Tabury K, Trittel T, Wehland M, Zhang YS, Putt KS, Zhang ZY, Tagle DA. Taking the 3Rs to a higher level: replacement and reduction of animal testing in life sciences in space research. Biotechnol Adv 2025; 81:108574. [PMID: 40180136 PMCID: PMC12048243 DOI: 10.1016/j.biotechadv.2025.108574] [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: 01/02/2025] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
Human settlements on the Moon, crewed missions to Mars and space tourism will become a reality in the next few decades. Human presence in space, especially for extended periods of time, will therefore steeply increase. However, despite more than 60 years of spaceflight, the mechanisms underlying the effects of the space environment on human physiology are still not fully understood. Animals, ranging in complexity from flies to monkeys, have played a pioneering role in understanding the (patho)physiological outcome of critical environmental factors in space, in particular altered gravity and cosmic radiation. The use of animals in biomedical research is increasingly being criticized because of ethical reasons and limited human relevance. Driven by the 3Rs concept, calling for replacement, reduction and refinement of animal experimentation, major efforts have been focused in the past decades on the development of alternative methods that fully bypass animal testing or so-called new approach methodologies. These new approach methodologies range from simple monolayer cultures of individual primary or stem cells all up to bioprinted 3D organoids and microfluidic chips that recapitulate the complex cellular architecture of organs. Other approaches applied in life sciences in space research contribute to the reduction of animal experimentation. These include methods to mimic space conditions on Earth, such as microgravity and radiation simulators, as well as tools to support the processing, analysis or application of testing results obtained in life sciences in space research, including systems biology, live-cell, high-content and real-time analysis, high-throughput analysis, artificial intelligence and digital twins. The present paper provides an in-depth overview of such methods to replace or reduce animal testing in life sciences in space research.
Collapse
Affiliation(s)
- Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Sarah Baatout
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium; Department of Molecular Biotechnology, Gent University, Gent, Belgium
| | - Bjorn Baselet
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Afshin Beheshti
- Center of Space Biomedicine, McGowan Institute for Regenerative Medicine, and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Markus Braun
- German Space Agency, German Aerospace Center, Bonn, Germany
| | | | - James A Casaletto
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Ben Cools
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium; Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Sylvain V Costes
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Phoebe De Meulemeester
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bartu Doruk
- Space Applications Services NV/SA, Sint-Stevens-Woluwe, Belgium; Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Silvana Miranda
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium; Department of Molecular Biotechnology, Gent University, Gent, Belgium
| | - Christiane Hahn
- European Space Agency, Human and Robotic Exploration Programmes, Human Exploration Science team, Noordwijk, the Netherlands
| | - Sinem Helvacıoğlu Akyüz
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Herbert
- Space Systems, Airbus Defence and Space, Immenstaad am Bodensee, Germany
| | - Dmitriy Krepkiy
- Office of Special Initiatives, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Yannick Lichterfeld
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Christian Liemersdorf
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Shannon Marchal
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jette Ritz
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Theresa Schmakeit
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Hilde Stenuit
- Space Applications Services NV/SA, Sint-Stevens-Woluwe, Belgium
| | - Kevin Tabury
- Nuclear Medical Applications Institute, Belgian Nuclear Research Centre, Mol, Belgium
| | - Torsten Trittel
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany; Department of Engineering, Brandenburg University of Applied Sciences, Brandenburg an der Havel, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University, Magdeburg, Germany
| | - Yu Shrike Zhang
- Division of Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Zhong-Yin Zhang
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA; Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Danilo A Tagle
- Office of Special Initiatives, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Rah B, Shafarin J, Qaisar R, Karim A, Hamad M, Muhammad JS. Mouse hindlimb unloading, as a model of simulated microgravity, leads to dysregulated iron homeostasis in liver and skeletal muscle cells. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:7-15. [PMID: 40280644 DOI: 10.1016/j.lssr.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 04/29/2025]
Abstract
Microgravity exposure can impact various physiological systems, yet its specific effects on liver cells remain inadequately studied. To address this gap, we used a hindlimb unloading (HU) mouse model to simulate microgravity conditions and investigate alterations in iron metabolism within liver and skeletal muscle cells. 16-week-old male C57BL/6j mice were divided into three groups: (i) ground-based control (GC), (ii) hindlimb unloading treated with vehicle (HU-v), and (iii) hindlimb unloading treated with deferoxamine (DFO). After three weeks, mice were euthanized, and samples of gastrocnemius muscle, liver, and serum were collected for analysis. The HU-v group exhibited significant muscle and liver cell atrophy compared to the GC group, along with disrupted iron metabolism, as indicated by altered expression of key iron regulatory proteins, including FTH1, FPN, TFR1, IRP-1, HMOX-1, and Hepcidin. In contrast, the DFO group demonstrated restored iron homeostasis, with protein expression patterns resembling those of the GC group. Serum analysis revealed elevated levels of serum iron, ferritin, and transferrin in the DFO group compared to both HU-v and GC groups, albeit with minimal changes in total iron-binding capacity. These findings suggest that simulated microgravity induces iron overload and cellular atrophy in liver and skeletal muscle cells, highlighting the potential therapeutic benefits of iron chelation in such conditions.
Collapse
Affiliation(s)
- Bilal Rah
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Jasmin Shafarin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Rizwan Qaisar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates
| | - Asima Karim
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, , United Arab Emirates; Department of Biomedical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
3
|
Morioka K, Tazoe T, Huie JR, Hayakawa K, Okazaki R, Guandique CF, Almeida CA, Haefeli J, Hamanoue M, Endoh T, Tanaka S, Bresnahan JC, Beattie MS, Ogata T, Ferguson AR. Disuse plasticity limits spinal cord injury recovery. iScience 2025; 28:112180. [PMID: 40224010 PMCID: PMC11987634 DOI: 10.1016/j.isci.2025.112180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Use-dependent plasticity after spinal cord injury (SCI) enhances neuromotor function, however, the optimal timing to initiate rehabilitation remains controversial. To test impacts of early disuse, we established a rodent model of transient hindlimb suspension in acute phase SCI. Early disuse in the first 2-week after SCI undermined recovery on open-field locomotion, kinematics, and swim tests even after 6-week of normal gravity reloading. Early disuse produced chronic spinal circuit hyper-excitability in H-reflex and interlimb reflex tests. Quantitative synaptoneurosome analysis of lumboventral spinal cords revealed shifts in AMPA receptor (AMPAR) subunit GluA1 localization and serine 881 phosphorylation, reflecting enduring synaptic memories of early disuse stored in the spinal cord. Automated confocal analysis of motoneurons revealed persistent shifts toward GluA2-lacking, calcium-permeable AMPARs in disuse subjects. Unsupervised machine learning associated multidimensional synaptic changes with persistent recovery deficits in SCI. The results argue for early aggressive rehabilitation to prevent disuse plasticity that limits SCI recovery.
Collapse
Affiliation(s)
- Kazuhito Morioka
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute (OTI), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Toshiki Tazoe
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Neural Prosthesis Project, Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - J. Russell Huie
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Kentaro Hayakawa
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Orthopaedic and Spine Surgery, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Rentaro Okazaki
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Orthopaedic Surgery, Saitama Red Cross Hospital, Saitama, Japan
| | - Cristian F. Guandique
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Carlos A. Almeida
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jenny Haefeli
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Makoto Hamanoue
- Department of Physiology, Advanced Medical Research Center, Toho University School of Medicine, Tokyo, Japan
| | - Takashi Endoh
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Faculty of Development and Education, Uekusa Gakuen University, Chiba, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jacqueline C. Bresnahan
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Michael S. Beattie
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Toru Ogata
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
- Department of Rehabilitation Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Adam R. Ferguson
- Department of Neurological Surgery, Weill Institute for Neurosciences, Brain and Spinal Injury Center (BASIC), University of California, San Francisco (UCSF), San Francisco, CA, USA
- San Francisco Veterans Affairs Healthcare System (SFVAHCS), San Francisco, CA, USA
| |
Collapse
|
4
|
Wuyts FL, Deblieck C, Vandevoorde C, Durante M. Brains in space: impact of microgravity and cosmic radiation on the CNS during space exploration. Nat Rev Neurosci 2025:10.1038/s41583-025-00923-4. [PMID: 40247135 DOI: 10.1038/s41583-025-00923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Solar system exploration is a grand endeavour of humankind. Space agencies have been planning crewed missions to the Moon and Mars for several decades. However, several environmental stress factors in space, such as microgravity and cosmic radiation, confer health risks for human explorers. This Review examines the effects of microgravity and exposure to cosmic radiation on the CNS. Microgravity presents challenges for the brain, necessitating the development of adaptive movement and orientation strategies to cope with alterations in sensory information. Exposure to microgravity also affects cognitive function to a certain extent. Recent MRI results show that microgravity affects brain structure and function. Post-flight recovery from these changes is gradual, with some lasting up to a year. Regarding cosmic radiation, animal experiments suggest that the brain could be much more sensitive to this stressor than may be expected from experiences on Earth. This may be due to the presence of energetic heavy ions in space that have an impact on cognitive function, even at low doses. However, all data about space radiation risk stem from rodent experiments, and extrapolation of these data to humans carries a high degree of uncertainty. Here, after presenting an overview of current knowledge in the above areas, we provide a concise description of possible counter-measures to protect the brain against microgravity and cosmic radiation during future space missions.
Collapse
Affiliation(s)
- Floris L Wuyts
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Choi Deblieck
- Laboratory for Equilibrium Investigations and Aerospace (LEIA), University of Antwerp, Antwerp, Belgium
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.
- Institute for Condensed Matter of Physics, Technische Universität Darmstadt, Darmstadt, Germany.
- Department of Physics 'Ettore Pancini', University Federico II, Naples, Italy.
| |
Collapse
|
5
|
Ranieri M, Venneri M, Storlino G, Ferrulli A, D’Agostino M, Centrone M, Di Mise A, Zerlotin R, Tamma G, Grano M, Valenti G. Alteration of vasopressin-aquaporin system in hindlimb unloading mice. Front Physiol 2025; 16:1535053. [PMID: 40303591 PMCID: PMC12037502 DOI: 10.3389/fphys.2025.1535053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Murine hindlimb unloading (HU) is considered a model of choice for simulating the physiological effects of microgravity on several functions, including fluid and electrolyte homeostasis. Microgravity causes changes in blood redistribution, modulating vasopressin secretion, a major hormone controlling water reabsorption through the vasopressin-sensitive water channel AQP2. In this study, mice were hindlimb suspended over 4 weeks or rested in the ground as controls, and vasopressin levels, along with renal aquaporins expression were investigated. Copeptin, a stable precursor of the hormone vasopressin, significantly increased as early as 1 week of unloading which correlated with a significant increase in AQP2 total protein expression and decrease in serum osmolality, suggesting early activation of the vasopressin/AQP2 axis in this model. Conversely, in 4 weeks HU suspended mice, copeptin decreased significantly and both AQP2 mRNA and AQP2 total protein expression were significantly reduced. Consistent with a downregulation of the vasopressin/AQP2 axis an increase in serum osmolality was observed at 4 weeks HU. The basolateral water channels AQP3 and AQP4 were, on the other hand, unaffected. Immunolocalization studies confirmed reduced expression of AQP2 in renal collecting ducts of HU mice at 4 weeks. A significantly increased amount of the expressed AQP2 was found phosphorylated at Ser261, a site regulating AQP2 protein stability and degradation. In line, p38-MAPK, committed to phosphorylate Ser261 and to increase miR137 expression, an AQP2 mRNA-targeted microRNA, was significantly increased in HU, suggesting that reduced AQP2 expression was mainly due to increased protein degradation and downregulation of AQP2-mRNA translation. Our results suggest that vasopressin/AQP2 axis is upregulated as early as 1 week and may be involved in the antidiuretic response also observed in early spaceflight period in astronauts. Contrariwise, the vasopressin-AQP2 system is downregulated after 4 weeks HU, likely to counteract the persistent central venous pressure due to cephalic shift of fluids.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Maria Venneri
- Istituti Clinici Scientifici Maugeri SPA SB IRCCS, Bari, Italy
| | - Giuseppina Storlino
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Angela Ferrulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Mariagrazia D’Agostino
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Bari, Italy
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Bari, Italy
| |
Collapse
|
6
|
Zhao H, Tu X. The potential key genes within focal adhesion that regulate mesenchymal stem cells osteogenesis or adipogenesis in microgravity related disuse osteoporosis: an integrated analysis. Front Endocrinol (Lausanne) 2025; 16:1469400. [PMID: 40130165 PMCID: PMC11930814 DOI: 10.3389/fendo.2025.1469400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
This study aimed to identify key genes related to focal adhesions (FA) and cells involved in osteoblast (OS) and adipocyte (AD) differentiation in osteoporosis. A mouse model of disuse osteoporosis was made by hindlimbs unloading (HLU)/Tail - suspension. Micro - CT and histological analysis were done, and differentially expressed genes (DEGs) from GSE100930 were analyzed. Soft clustering on GSE80614 OS/AD samples found FA - related candidate genes. protein-protein interaction (PPI) network and cytoHubba's Degree algorithm identified key FA - genes, validated by quantitative polymerase chain reaction (qPCR). Key OS/AD - associated cells were identified by single - cell analysis. The mouse model showed decreased bone density, microstructure damage, increased marrow adiposity, and altered gene expression. Key FA - related genes for osteogenesis (ITGB3, LAMC1, COL6A3, ITGA8, PDGFRB) and adipogenesis (ITGB3, ITGA4, LAMB1, ITGA8, LAMA4) were found and validated. Key cells (chondrocyte, adipocyte, and osteoblast progenitors) are involved in specific pathways, with osteoblast progenitors having stronger interactions. Pseudotime analysis implies differentiation from chondrocyte progenitors to adipocyte, then osteoblast progenitors. This study provides new insights for disuse osteoporosis research.
Collapse
Affiliation(s)
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Galdamez LA, Mader TH, Ong J, Kadipasaoglu CM, Lee AG. A multifactorial, evidence-based analysis of pathophysiology in Spaceflight Associated Neuro-Ocular Syndrome (SANS). Eye (Lond) 2025; 39:700-709. [PMID: 39827235 PMCID: PMC11885454 DOI: 10.1038/s41433-025-03618-3] [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: 12/16/2023] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
The National Aeronautics and Space Administration (NASA) in the United States has been studying a fascinating and unique constellation of neuro-ophthalmic findings collectively known as Spaceflight Associated Neuro-Ocular Syndrome (SANS). SANS is unique to the space environment of microgravity and produces novel physiological and pathological findings that have no direct terrestrial equivalent. The neuro-ophthalmic phenomenon is a major physiologic barrier to future planetary spaceflight. The underlying pathophysiology of SANS remains ill-defined, but since its initial report in 2011, several hypotheses have been proposed including increased intracranial pressure, cerebral venous congestion and glymphatic stasis, compartmentalization of CSF within the orbital nerve sheath sub-arachnoid space (SAS), upward brain shift, inflammation, disrupted axoplasmic transport, and radiation exposure. These aetiologies may not be mutually exclusive and may be interconnected, leading to an integrative, multifactorial aetiology of SANS. This paper critically analyses the various hypotheses of this neuro-ophthalmic phenomenon and the connections between the physiologic and anatomical evidence-based changes observed in spaceflight and terrestrial analogues. Continued prospective, longitudinal study and development of practical countermeasures for SANS will be necessary for future human spaceflight missions including the mission to Mars.
Collapse
Affiliation(s)
- Laura A Galdamez
- Department of Emergency Medicine, Memorial Hermann The Woodlands, The Woodlands, TX, USA
| | | | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | | | - Andrew G Lee
- Department of Ophthalmology, Houston Methodist Hospital, Houston, TX, USA.
- Baylor College of Medicine and the Baylor Center for Space Medicine, Houston, TX, USA.
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine New York, New York, NY, USA.
- Department of Ophthalmology, The University of Texas Medical Branch (UTMB), Galveston, TX, USA.
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- The UT MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Zhang X, Zhu H, Zhang J. Oxidative Stress on the Ground and in the Microgravity Environment: Pathophysiological Effects and Treatment. Antioxidants (Basel) 2025; 14:231. [PMID: 40002415 PMCID: PMC11852023 DOI: 10.3390/antiox14020231] [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: 01/23/2025] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
With the continued exploration of the universe, there is an increasingly urgent need to address the health challenges arising from spaceflight. In space, astronauts are exposed to radiation, confinement and isolation, circadian rhythm dysregulation, and microgravity conditions that are different from those on Earth. These risk factors jeopardize astronauts' health, thus affecting the quality of space missions. Among these factors, gravitational changes influence the balance between oxidation and antioxidants, stimulating the production of reactive oxygen species (ROS), finally leading to oxidative stress (OS). OS leads to oxidative damage of biomolecules such as lipids, proteins, and DNA, which causes the development of various diseases. The occurrence of OS is increased in microgravity and affects multiple systems, including the musculoskeletal, cardiovascular, nervous, and immune systems. In this review, we discuss the mechanisms of OS, the physiological effects on different systems caused by OS in microgravity environment, and potential treatments for OS. Finally, treatment strategies for oxidative stress in microgravity are summarized, providing some promising approaches for protecting the health of astronauts in future space exploration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (X.Z.); (H.Z.)
| | - Huaiying Zhu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (X.Z.); (H.Z.)
| | - Jinhua Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (X.Z.); (H.Z.)
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
9
|
Chiaberge M, Thottappillil N, Liphardt AM, Furlanetto A, Odell D, Wang C, Hope S, Smee S, Rehfus J, Niehoff A, Shelhamer M, Norman C, Philippon MJ, Huard J, James AW, Fan CM. Plyometric training increases thickness and volume of knee articular cartilage in mice. NPJ Microgravity 2025; 11:5. [PMID: 39948108 PMCID: PMC11825961 DOI: 10.1038/s41526-025-00458-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Degeneration and thinning of articular cartilage lead to osteoarthritis and may result from reduced joint loading during e.g. bed rest or as a result of microgravity during space flight. Anabolic physical exercises for cartilage are not well studied to date. We built an experimental apparatus for plyometric training with mice to test potential benefits of jumping for articular cartilage. The exercise group (JUMP) performed jump training for 9 weeks and was compared with sedentary mice (control, CON) and hindlimb-suspended (HLS) mice (to simulate reduced loading) for the same duration. Knee cartilage was assessed via 3-dimensional reconstruction of micro-CT scans and histology. We observed significant thinning and volume reduction of articular cartilage at the medial tibial-femoral point of contact in the HLS group. Clustering of chondrocytes was present in HLS. By contrast, the JUMP group showed both cartilage thickening and volume increase. We observed a similar trend on trabecular bone thickness and volume. Our results show that plyometric training can stimulate cartilage thickness and volume in mice. This suggests further investigation of this mode of exercise as a countermeasure to prevent cartilage atrophy in disuse scenarios such as long duration spaceflight, and for patients at risk of developing osteoarthritis.
Collapse
Affiliation(s)
- Marco Chiaberge
- The William H. Miller III Department of Physics & Astronomy, Johns Hopkins University, Baltimore, MD, USA.
- Space Telescope Science Institute for the European Space Agency (ESA), ESA Office, 3700 San Martin Drive, Baltimore, MD, USA.
| | - Neelima Thottappillil
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna-Maria Liphardt
- Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Anderson Furlanetto
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Dylan Odell
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Christine Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen Hope
- The William H. Miller III Department of Physics & Astronomy, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen Smee
- The William H. Miller III Department of Physics & Astronomy, Johns Hopkins University, Baltimore, MD, USA
| | - Joseph Rehfus
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany
- Cologne Center for Musculoskeletal Biomechanics, University of Cologne, Faculty of Medicine, Köln, Germany
| | - Mark Shelhamer
- Human Spaceflight Lab, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Colin Norman
- The William H. Miller III Department of Physics & Astronomy, Johns Hopkins University, Baltimore, MD, USA
- Space Telescope Science Institute for the European Space Agency (ESA), ESA Office, 3700 San Martin Drive, Baltimore, MD, USA
| | - Marc J Philippon
- Steadman Philippon Research Institute, Vail, CO, USA
- The Steadman Clinic, Vail, CO, USA
| | - Johnny Huard
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chen-Ming Fan
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
| |
Collapse
|
10
|
Zheng Y, Yu Y, Chen M, Zhang H, Wang W, Fan X, Sun L, Tang L, Ta D. Abdominal LIPUS Stimulation Prevents Cognitive Decline in Hind Limb Unloaded Mice by Regulating Gut Microbiota. Mol Neurobiol 2025:10.1007/s12035-025-04709-8. [PMID: 39878866 DOI: 10.1007/s12035-025-04709-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
Weightlessness usually causes disruption of the gut microbiota and impairs cognitive function. There is a close connection between gut microbiota and neurological diseases. Low-intensity pulsed ultrasound (LIPUS) has a beneficial effect on reducing intestinal inflammation. So we wondered if abdominal LIPUS stimulation can have a positive impact on weightlessness induced cognitive decline by reducing intestinal dysfunction. The findings revealed that the hind limb unloaded mice exhibited evident disruption in intestinal structure and gut microbial homeostasis, along with impairment in their learning and memory capabilities. However, 4-week abdominal LIPUS treatment improved intestinal function in hind limb unloaded mice, characterized by upregulation of tight junction proteins ZO-1 and Occludin expression in the colon, increased diversity and abundance of intestinal microbiota, decreased serum lipopolysaccharide (LPS), and increased short chain fatty acids in colon contents. The hind limb unloaded mice treated with LIPUS exhibited heightened activity levels, improved exploratory tendencies, and significantly enhanced learning and memory faculties, and elevated expression of neuroadaptation-related proteins such as PSD95, GAP43, P-CREB, BDNF, and its receptor TRKB in the hippocampus. Furthermore, the hind limb unloaded mice receiving fecal transplants from the mice whose abdomens were irradiated with LIPUS displayed enhanced cognitive abilities and improved intestinal structure, akin to the outcomes observed in hind limb unloaded mice who received LIPUS abdominal treatment directly. The above results indicate that LIPUS enhances intestinal structure and microbiota, which helps alleviate cognitive impairment caused by weightlessness. LIPUS could be a potential strategy to simultaneously improve gut dysfunction and cognitive decline in astronauts or bedridden patients.
Collapse
Affiliation(s)
- Yumei Zheng
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengyao Chen
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Huiyuan Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Wanzhao Wang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, 710119, China.
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, 200433, China.
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
11
|
Muramatsu W, Maryanovich M, Akiyama T, Karagiannis GS. Thymus ad astra, or spaceflight-induced thymic involution. Front Immunol 2025; 15:1534444. [PMID: 39926601 PMCID: PMC11802524 DOI: 10.3389/fimmu.2024.1534444] [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: 11/25/2024] [Accepted: 12/24/2024] [Indexed: 02/11/2025] Open
Abstract
Spaceflight imposes a constellation of physiological challenges-cosmic radiation, microgravity, disrupted circadian rhythms, and psychosocial stress-that critically compromise astronaut health. Among the most vulnerable organs is the thymus, a cornerstone of immune system functionality, tasked with generating naive T cells essential for adaptive immunity. The thymus is particularly sensitive to spaceflight conditions, as its role in maintaining immune homeostasis is tightly regulated by a balance of systemic and local factors easily disrupted in space. Cosmic radiation, an omnipresent hazard beyond Earth's magnetosphere, accelerates DNA damage and cellular senescence in thymic epithelial cells, impairing thymopoiesis and increasing the risk of immune dysregulation. Microgravity and circadian rhythm disruption exacerbate this by altering immune cell migration patterns and stromal support, critical for T-cell development. Psychosocial stressors, including prolonged isolation and mission-induced anxiety, further compound thymic atrophy by elevating systemic glucocorticoid levels. Ground-based analogs simulating cosmic radiation and microgravity have been instrumental in elucidating mechanisms of thymic involution and its downstream effects on immunity. These models reveal that long-duration missions result in diminished naive T-cell output, leaving astronauts vulnerable to infections and possibly at high risk for developing neoplasia. Advances in countermeasures, such as pharmacological interventions targeting thymic regeneration and bioengineering approaches to protect thymic architecture, are emerging as vital strategies to preserve immune resilience during prolonged space exploration. Focusing on the thymus as a central hub of immune vulnerability underscores its pivotal role in spaceflight-induced health risks. Understanding these dynamics will not only enhance the safety of human space missions but also provide critical insights into thymus biology under extreme conditions.
Collapse
Affiliation(s)
- Wataru Muramatsu
- Laboratory of Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
- Cancer Dormancy Institute, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
| | - Taishin Akiyama
- Laboratory of Immune Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - George S. Karagiannis
- Cancer Dormancy Institute, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, United States
- The Marilyn and Stanely M. Katz Institute for Immunotherapy for Cancer and Inflammatory Disorders, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
| |
Collapse
|
12
|
Elsangeedy E, Yamaleyeva DN, Edenhoffer NP, Deak A, Soloshenko A, Ray J, Sun X, Shaltout OH, Cruz-Diaz N, Westwood B, Kim-Shapiro D, Diz DI, Soker S, Pulgar VM, Ronca A, Willey JS, Yamaleyeva LM. Sex-specific cardiovascular adaptations to simulated microgravity in Sprague-Dawley rats. NPJ Microgravity 2024; 10:110. [PMID: 39702444 DOI: 10.1038/s41526-024-00450-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024] Open
Abstract
Men and women have different cardiovascular responses to spaceflight; however, few studies have focused on direct comparisons between sexes. We investigated the mechanisms of aortic stiffening in socially and sexually mature 20-week-old male and female Sprague Dawley (SD) rats exposed to hindlimb unloading (HLU) for 14 days. Pulse wave velocity (PWV) was greater in the aortic arch of females after HLU versus control females (n = 6-8). HLU had no effect on aortic PWV in males (n = 5-6). Aortic α smooth muscle actin, myosin, collagen, elastin, and collagen-to-elastin ratio were not different in rats of either sex following HLU. The levels of G protein-coupled estrogen receptor (GPER) were lower in the aorta of SD females exposed to HLU compared with female controls but were not altered in males. HLU females also had lower aortic PPARγ, increased oxidative stress markers, and diastolic dysfunction compared with control females. GPER agonist G1 prevented the increase in PWV and 8-hydroxy-2'-deoxyguanosine without altering PPARγ or p47phox in HLU females (n = 4 in each group) suggesting that lower GPER may contribute to arterial stiffening in the setting of simulated microgravity. This study highlights sex-specific vascular adaptations to the state of simulated microgravity.
Collapse
Affiliation(s)
- Ebrahim Elsangeedy
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dina N Yamaleyeva
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Nicholas P Edenhoffer
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Allyson Deak
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anna Soloshenko
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jonathan Ray
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Xuming Sun
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Omar H Shaltout
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Nildris Cruz-Diaz
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Brian Westwood
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Debra I Diz
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Victor M Pulgar
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pharmaceutical & Clinical Sciences, Campbell University, Buies Creek, NC, USA
| | - April Ronca
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, USA
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Liliya M Yamaleyeva
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
13
|
Zhang X, Zhou X, Tu Z, Qiang L, Lu Z, Xie Y, Liu CH, Zhang L, Fu Y. Proteomic and ubiquitinome analysis reveal that microgravity affects glucose metabolism of mouse hearts by remodeling non-degradative ubiquitination. PLoS One 2024; 19:e0313519. [PMID: 39541295 PMCID: PMC11563481 DOI: 10.1371/journal.pone.0313519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Long-term exposure to a microgravity environment leads to structural and functional changes in hearts of astronauts. Although several studies have reported mechanisms of cardiac damage under microgravity conditions, comprehensive research on changes at the protein level in these hearts is still lacking. In this study, proteomic analysis of microgravity-exposed hearts identified 156 differentially expressed proteins, and ubiquitinomic analysis of these hearts identified 169 proteins with differential ubiquitination modifications. Integrated ubiquitinomic and proteomic analysis revealed that differential proteomic changes caused by transcription affect the immune response in microgravity-exposed hearts. Additionally, changes in ubiquitination modifications under microgravity conditions excessively activated certain kinases, such as hexokinase and phosphofructokinase, leading to cardiac metabolic disorders. These findings provide new insights into the mechanisms of cardiac damage under microgravity conditions.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xuemei Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhiwei Tu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lihua Qiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhe Lu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuping Xie
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Cui Hua Liu
- Institute of Microbiology (Chinese Academy of Sciences), CAS Key Laboratory of Pathogenic Microbiology and Immunology, Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yesheng Fu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
14
|
Zhang S, Zhang S, Wang Z, Adachi T, Yoshida Y, Takahashi A. Disparity in the effect of partial gravity simulated using a new apparatus on different rat hindlimb muscles. LIFE SCIENCES IN SPACE RESEARCH 2024; 43:54-67. [PMID: 39521495 DOI: 10.1016/j.lssr.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 11/16/2024]
Abstract
The days of returning to the Moon and landing on Mars are approaching. These long-duration missions present significant challenges, such as changes in gravity, which pose serious threats to human health. Maintaining muscle function and health is essential for successful spaceflight and exploration of the Moon and Mars. This study aimed to observe the adaptation of rat hindlimb muscles to partial gravity conditions by simulating the gravity of space (microgravity (µG)), Moon (1/6G), and Mars (3/8G) using our recently invented ground-based apparatus. A total of 25 rats were included in this study. The rats were divided into five groups: control (1G), sham (1G), simulated Mars (3/8G), simulated Moon (1/6G), and simulated Space (µG). Muscle mass, fiber proportion, and fiber cross-sectional area (CSA) of four types of hindlimb muscles were measured: gastrocnemius (GA), tibialis anterior (TA), extensor digitorum longus (EDL), soleus (Sol). Sol and GA exhibited the most significant alterations in response to the changes in gravity after 10 days of the experiment. A notable decline in muscle mass was observed in the simulated µG, Moon, and Mars groups, with the µG group exhibiting the most noticeable decline. In Sol, a noteworthy decline in the proportion of slow-twitch type I fibers, CSA of slow-twitch type I fibers, and average CSA of the whole muscle fibers was observed in the simulated groups. The GA red, mixed, and white portions were examined, and the GA mixed portion showed significant differences in fiber proportion and CSA. A notable increase in the proportion of slow-twitch type I fibers was observed in the simulated groups, with a significant decrease in CSA of type IIb. In EDL or TA, no discernible changes in muscle mass, fiber proportion, or fiber CSA were observed in any of the five groups. These findings indicate that weight-bearing muscles, such as Sol and GA, are more sensitive to changes in partial gravity. Furthermore, partial gravity is insufficient to preserve the normal physiological and functional properties of the hindlimb muscles. Therefore, targeted muscle interventions are required to ensure astronauts' health and mission success. Furthermore, these findings demonstrate the viability and durability of our ground-based apparatus for partial gravity simulation.
Collapse
Affiliation(s)
- Shengli Zhang
- Graduate School of Medicine Medical Sciences, Gunma University, Maebashi, Gunma 371-8511, Japan.
| | - Shenke Zhang
- Graduate School of Medicine Medical Sciences, Gunma University, Maebashi, Gunma 371-8511, Japan.
| | - Zhen Wang
- Division of Biology, Gunma University Heavy Ion Medical Center, Maebashi, Gunma 371-8511, Japan.
| | - Takuya Adachi
- Graduate School of Medicine Medical Sciences, Gunma University, Maebashi, Gunma 371-8511, Japan.
| | - Yukari Yoshida
- Division of Biology, Gunma University Heavy Ion Medical Center, Maebashi, Gunma 371-8511, Japan.
| | - Akihisa Takahashi
- Division of Biology, Gunma University Heavy Ion Medical Center, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
15
|
Han H, Jia H, Wang YF, Song JP. Cardiovascular adaptations and pathological changes induced by spaceflight: from cellular mechanisms to organ-level impacts. Mil Med Res 2024; 11:68. [PMID: 39334239 PMCID: PMC11429428 DOI: 10.1186/s40779-024-00570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
The advancement in extraterrestrial exploration has highlighted the crucial need for studying how the human cardiovascular system adapts to space conditions. Human development occurs under the influence of gravity, shielded from space radiation by Earth's magnetic field, and within an environment characterized by 24-hour day-night cycles resulting from Earth's rotation, thus deviating from these conditions necessitates adaptive responses for survival. With upcoming manned lunar and Martian missions approaching rapidly, it is essential to understand the impact of various stressors induced by outer-space environments on cardiovascular health. This comprehensive review integrates insights from both actual space missions and simulated experiments on Earth, to analyze how microgravity, space radiation, and disrupted circadian affect cardiovascular well-being. Prolonged exposure to microgravity induces myocardial atrophy and endothelial dysfunction, which may be exacerbated by space radiation. Mitochondrial dysfunction and oxidative stress emerge as key underlying mechanisms along with disturbances in ion channel perturbations, cytoskeletal damage, and myofibril changes. Disruptions in circadian rhythms caused by factors such as microgravity, light exposure, and irregular work schedules, could further exacerbate cardiovascular issues. However, current research tends to predominantly focus on disruptions in the core clock gene, overlooking the multifactorial nature of circadian rhythm disturbances in space. Future space missions should prioritize targeted prevention strategies and early detection methods for identifying cardiovascular risks, to preserve astronaut health and ensure mission success.
Collapse
Affiliation(s)
- Han Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yi-Fan Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiang-Ping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
16
|
Hao R, Tang H, Ding C, Rajbanshi B, Liu Y, Ma D, Duan Z, Qi Y, Dai L, Zhang B, Zhang A, Zhang X. A Novel Piezo1 Agonist Promoting Mesenchymal Stem Cell Proliferation and Osteogenesis to Attenuate Disuse Osteoporosis. SMALL SCIENCE 2024; 4:2400061. [PMID: 40212079 PMCID: PMC11935128 DOI: 10.1002/smsc.202400061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/12/2024] [Indexed: 04/13/2025] Open
Abstract
Disuse osteoporosis (OP) is a state of bone loss due to lack of mechanical stimuli, probably induced by prolonged bed rest, neurological diseases, as well as microgravity. Currently the precise treatment strategies of disuse OP remain largely unexplored. Piezo1, a mechanosensitive calcium (Ca2+) ion channel, is a key force sensor mediating mechanotransduction and it is demonstrated to regulate bone homeostasis and osteogenesis in response to mechanical forces. Using structure-based drug design, a novel small-molecule Piezo1 agonist, MCB-22-174, which can effectively activate Piezo1 and initiate Ca2+ influx, is developed and is more potent than the canonical Piezo1 agonist, Yoda1. Moreover, MCB-22-174 is found as a safe Piezo1 agonist without any signs of serious toxicity. Mechanistically, Piezo1 activation promotes the proliferation of bone marrow mesenchymal stem cells by activating the Ca2+-related extracellular signal-related kinases and calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) pathway. Importantly, MCB-22-174 could effectively promote osteogenesis and attenuate disuse OP in vivo. Overall, the findings provide a promising therapeutic strategy for disuse OP by chemical activation of Piezo1.
Collapse
Affiliation(s)
- Ruihan Hao
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Hairong Tang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Chunyong Ding
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Bhavana Rajbanshi
- Department of Dermatology and VenereologyTongji University School of MedicineShanghai200092China
| | - Yuhang Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Ding Ma
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Zhouyi Duan
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Yuxin Qi
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and MinistryGuangxi Medical UniversityNanningGuangxi530021China
| | - Liming Dai
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Ao Zhang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| |
Collapse
|
17
|
Ye R, He Y, Ni W, Zhang Y, Zhu Y, Cao M, He R, Yao M. LLLT accelerates experimental wound healing under microgravity conditions via PI3K/AKT-CCR2 signal axis. Front Bioeng Biotechnol 2024; 12:1387474. [PMID: 39193227 PMCID: PMC11347831 DOI: 10.3389/fbioe.2024.1387474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Background and Purpose The risk of skin injuries in space is increasing with longer space missions and a growing astronaut population. This highlights the importance of understanding the adverse effects of weightlessness on wound healing. The objective of this research was to examine the therapeutic potential of Low-Level Light Therapy (LLLT) on skin healing processes under simulated microgravity (SMG) conditions and uncover the underlying molecular mechanisms, thus providing innovative solutions and a sound theoretical basis for space skin injuries. Methods Hindlimb unloading (HU) mice models were used to simulate weightlessness conditions, with or without a complete management of LLLT for 14 days. A systematic testing consisting of HE, Masson and immunohistochemical staining was performed against the standardized mouse tissue specimens. In vitro assessment of cellular biological functions under SMG conditions was carried out in the rotation system of culture (RSOC) using HaCaT and NIH3T3 cell-lines. Results Under SMG conditions, LLLT significantly reduced skin wound area in HU mice, especially on Days 10 (p < 0.001), accompanied by increased collagen deposition and elevated levels of Ki67 and CD31. Moreover, LLLT showed impressive anti-inflammatory effects represented by the reduced in pro-inflammatory markers including LY6G, F4/80 and CD86, as well as the decreased levels of IL-1β, IL-6 and TNF-α. Conversely, an elevation in the anti-inflammatory marker CD206 was observed. By employing bioinformatics technology, we further found the PI3K/AKT signaling was prominent in the KEGG pathway analysis and CCR2 acted as a hub gene in the interaction network. Therefore, we demonstrated that LLLT could enhance the phosphorylation of PI3K/AKT and reduce CCR2 expression under SMG conditions, while CCR2 knockdown promoted the phosphorylation of PI3K/AKT, suggesting an important role of CCR2/PI3K/AKT signal axis in LLLT-accelerated wound healing under SMG conditions. Conclusion LLLT induced activation of the PI3K/AKT signaling pathway through suppression of CCR2 expression, which significantly enhanced skin wound healing under SMG conditions.s.
Collapse
Affiliation(s)
- Rongan Ye
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yu He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Wei Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yiqiu Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ying Zhu
- Shanghai Key Laboratory of Reproductive Medicine, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Muqing Cao
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Ruida He
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Zong B, Wang J, Wang K, Hao J, Han JY, Jin R, Ge Q. Effects of Ginsenoside Rb1 on the Crosstalk between Intestinal Stem Cells and Microbiota in a Simulated Weightlessness Mouse Model. Int J Mol Sci 2024; 25:8769. [PMID: 39201456 PMCID: PMC11354315 DOI: 10.3390/ijms25168769] [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: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Exposure to the space microenvironment has been found to disrupt the homeostasis of intestinal epithelial cells and alter the composition of the microbiota. To investigate this in more detail and to examine the impact of ginsenoside Rb1, we utilized a mouse model of hindlimb unloading (HU) for four weeks to simulate the effects of microgravity. Our findings revealed that HU mice had ileum epithelial injury with a decrease in the number of intestinal stem cells (ISCs) and the level of cell proliferation. The niche functions for ISCs were also impaired in HU mice, including a reduction in Paneth cells and Wnt signaling, along with an increase in oxidative stress. The administration of Rb1 during the entire duration of HU alleviated the observed intestinal defects, suggesting its beneficial influence on epithelial cell homeostasis. Hindlimb unloading also resulted in gut dysbiosis. The supplementation of Rb1 in the HU mice or the addition of Rb1 derivative compound K in bacterial culture in vitro promoted the growth of beneficial probiotic species such as Akkermansia. The co-housing experiment further showed that Rb1 treatment in ground control mice alone could alleviate the defects in HU mice that were co-housed with Rb1-treated ground mice. Together, these results underscore a close relationship between dysbiosis and impaired ISC functions in the HU mouse model. It also highlights the beneficial effects of Rb1 in mitigating HU-induced epithelial injury by promoting the expansion of intestinal probiotics. These animal-based insights provide valuable knowledge for the development of improved approaches to maintaining ISC homeostasis in astronauts.
Collapse
Affiliation(s)
- Beibei Zong
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (B.Z.); (J.-Y.H.)
| | - Jingyi Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (J.W.); (K.W.); (J.H.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (J.W.); (K.W.); (J.H.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Jie Hao
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (J.W.); (K.W.); (J.H.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (B.Z.); (J.-Y.H.)
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (J.W.); (K.W.); (J.H.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| | - Qing Ge
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (B.Z.); (J.-Y.H.)
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (J.W.); (K.W.); (J.H.)
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China
| |
Collapse
|
19
|
Gao M, Dong C, Chen Z, Jiang R, Shaw P, Gao W, Sun Y. Different impact of short-term and long-term hindlimb disuse on bone homeostasis. Gene 2024; 918:148457. [PMID: 38641071 DOI: 10.1016/j.gene.2024.148457] [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: 11/14/2023] [Revised: 02/19/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
Disuse osteoporosis is one of the major problems of bone health which commonly occurs in astronauts during long-term spaceflight and bedridden patients. However, the mechanisms underlying such mechanical unloading induced bone loss have not been fully understood. In this study, we employed hindlimb-unloading mice models with different length of tail suspension to investigate if the bone loss was regulated by distinct factors under different duration of disuse. Our micro-CT results showed more significant decrease of bone mass in 6W (6-week) tail-suspension mice compared to the 1W (1-week) tail-suspension ones, as indicated by greater reduction of BV/TV, Tb.N, B.Ar/T.Ar and Ct.Th. RNA-sequencing results showed significant effects of hindlimb disuse on cell locomotion and immune system process which could cause bone loss.Real-time quantitative PCR results indicated a greater number of bone formation related genes that were downregulated in short-term tail-suspension mice compared to the long-term ones. It is, thus, suggested while sustained hindlimb unloading continuously contributes to bone loss, molecular regulation of bone homeostasis tends to reach a balance during this process.
Collapse
Affiliation(s)
- Minhao Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengji Dong
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhuliu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Renhao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peter Shaw
- Oujiang Lab, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang 325000, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yuanna Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
20
|
Zhang Y, Zhan Y, Guo K, Zheng Y, Tang L, Guo J, Liang J. Contrastive Disentanglement for Quantitative Ultrasound Muscle Atrophy Evaluation. IEEE Trans Biomed Eng 2024; 71:2352-2366. [PMID: 38393845 DOI: 10.1109/tbme.2024.3369072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
OBJECTIVE Muscleatrophy reduces the quality of life and increases morbidity and mortality from other diseases. The development of non-invasive muscle atrophy evaluation method is of great practical value. The lack of gold standard for pathological grading usually allows only the duration of weightlessness as a criterion for the degree of atrophy. However, the adaptive reductive remodeling of muscle physiology and structure shows a trend of nonlinear changes in time. Consequently, using weightlessness time as a benchmark for the degree of atrophy is inaccurate. METHODS This paper proposes a new ultrasound imaging-based method for quantifying muscle atrophy that utilizes weakly supervised information between multiple data partitions with controlled variance components, overcoming the limitations of using the weightlessness time as a criterion. We introduce a group-supervised contrastive disentanglement network (GCDNet) to disentangle the individual variances, muscle growth and atrophy components of ultrasound images, and quantify the degree of atrophy using the disentangled atrophy component. RESULTS The feasibility of GCDNet is validated by the separability, independence, and representativeness of the disentangled components. To simplify the application of GCDNet, a muscle atrophy scoring network requiring no reference images is developed by distilling the GCDNet's knowledge of muscle atrophy quantization. The strength of the proposed methodology allows us, for the first time to our knowledge, to study the muscle growth attribute during hind-limb unloading and the spatial distribution of muscle atrophy.
Collapse
|
21
|
Tahimic CGT, Steczina S, Sebastian A, Hum NR, Abegaz M, Terada M, Cimini M, Goukassian DA, Schreurs AS, Hoban-Higgins TM, Fuller CA, Loots GG, Globus RK, Shirazi-Fard Y. Simulated Microgravity Alters Gene Regulation Linked to Immunity and Cardiovascular Disease. Genes (Basel) 2024; 15:975. [PMID: 39202335 PMCID: PMC11353732 DOI: 10.3390/genes15080975] [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: 06/29/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/03/2024] Open
Abstract
Microgravity exposure induces a cephalad fluid shift and an overall reduction in physical activity levels which can lead to cardiovascular deconditioning in the absence of countermeasures. Future spaceflight missions will expose crew to extended periods of microgravity among other stressors, the effects of which on cardiovascular health are not fully known. In this study, we determined cardiac responses to extended microgravity exposure using the rat hindlimb unloading (HU) model. We hypothesized that exposure to prolonged simulated microgravity and subsequent recovery would lead to increased oxidative damage and altered expression of genes involved in the oxidative response. To test this hypothesis, we examined hearts of male (three and nine months of age) and female (3 months of age) Long-Evans rats that underwent HU for various durations up to 90 days and reambulated up to 90 days post-HU. Results indicate sex-dependent changes in oxidative damage marker 8-hydroxydeoxyguanosine (8-OHdG) and antioxidant gene expression in left ventricular tissue. Three-month-old females displayed elevated 8-OHdG levels after 14 days of HU while age-matched males did not. In nine-month-old males, there were no differences in 8-OHdG levels between HU and normally loaded control males at any of the timepoints tested following HU. RNAseq analysis of left ventricular tissue from nine-month-old males after 14 days of HU revealed upregulation of pathways involved in pro-inflammatory signaling, immune cell activation and differential expression of genes associated with cardiovascular disease progression. Taken together, these findings provide a rationale for targeting antioxidant and immune pathways and that sex differences should be taken into account in the development of countermeasures to maintain cardiovascular health in space.
Collapse
Affiliation(s)
- Candice G. T. Tahimic
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
| | - Sonette Steczina
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
- Blue Marble Space Institute of Science, Seattle, WA 98104, USA
| | - Aimy Sebastian
- Lawrence Livermore National Laboratory, Livermore, CA 94550, USA (G.G.L.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Livermore, CA 94550, USA (G.G.L.)
| | - Metadel Abegaz
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
- Blue Marble Space Institute of Science, Seattle, WA 98104, USA
| | - Masahiro Terada
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
- Universities Space Research Association, Washington, DC 20024, USA
| | - Maria Cimini
- Temple University School of Medicine, Philadelphia, PA 19140, USA;
| | - David A. Goukassian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
- Universities Space Research Association, Washington, DC 20024, USA
| | - Tana M. Hoban-Higgins
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Charles A. Fuller
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA 95616, USA
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Livermore, CA 94550, USA (G.G.L.)
- Department of Orthopedic Surgery, University of California Davis Health, Sacramento, CA 95817, USA
| | - Ruth K. Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
| | - Yasaman Shirazi-Fard
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA (M.A.); (Y.S.)
| |
Collapse
|
22
|
Gros A, Furlan FM, Rouglan V, Favereaux A, Bontempi B, Morel JL. Physical exercise restores adult neurogenesis deficits induced by simulated microgravity. NPJ Microgravity 2024; 10:69. [PMID: 38906877 PMCID: PMC11192769 DOI: 10.1038/s41526-024-00411-6] [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: 11/03/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Cognitive impairments have been reported in astronauts during spaceflights and documented in ground-based models of simulated microgravity (SMG) in animals. However, the neuronal causes of these behavioral effects remain largely unknown. We explored whether adult neurogenesis, known to be a crucial plasticity mechanism supporting memory processes, is altered by SMG. Adult male Long-Evans rats were submitted to the hindlimb unloading model of SMG. We studied the proliferation, survival and maturation of newborn cells in the following neurogenic niches: the subventricular zone (SVZ)/olfactory bulb (OB) and the dentate gyrus (DG) of the hippocampus, at different delays following various periods of SMG. SMG exposure for 7 days, but not shorter periods of 6 or 24 h, resulted in a decrease of newborn cell proliferation restricted to the DG. SMG also induced a decrease in short-term (7 days), but not long-term (21 days), survival of newborn cells in the SVZ/OB and DG. Physical exercise, used as a countermeasure, was able to reverse the decrease in newborn cell survival observed in the SVZ and DG. In addition, depending on the duration of SMG periods, transcriptomic analysis revealed modifications in gene expression involved in neurogenesis. These findings highlight the sensitivity of adult neurogenesis to gravitational environmental factors during a transient period, suggesting that there is a period of adaptation of physiological systems to this new environment.
Collapse
Affiliation(s)
- Alexandra Gros
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Centre National d'Etudes Spatiales, F-75001, Paris, France
| | - Fandilla Marie Furlan
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
- Department of Genetics & Evolution, 30 Quai Ernest-Ansermet, 1205, Geneva, Switzerland
| | - Vanessa Rouglan
- CNRS, IINS, UMR 5297, University Bordeaux, F-33000, Bordeaux, France
| | | | - Bruno Bontempi
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France
| | - Jean-Luc Morel
- CNRS, INCIA, UMR 5287, University Bordeaux, F-33000, Bordeaux, France.
- CNRS, IMN, UMR 5293, University Bordeaux, F-33000, Bordeaux, France.
| |
Collapse
|
23
|
An R, Blackwell VK, Harandi B, Gibbons AC, Siu O, Irby I, Rees A, Cornejal N, Sattler KM, Sheng T, Syracuse NC, Loftus D, Santa Maria SR, Cekanaviciute E, Reinsch SS, Ray HE, Paul AM. Influence of the spaceflight environment on macrophage lineages. NPJ Microgravity 2024; 10:63. [PMID: 38862517 PMCID: PMC11166655 DOI: 10.1038/s41526-023-00293-0] [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: 11/27/2022] [Accepted: 05/25/2023] [Indexed: 06/13/2024] Open
Abstract
Spaceflight and terrestrial spaceflight analogs can alter immune phenotypes. Macrophages are important immune cells that bridge the innate and adaptive immune systems and participate in immunoregulatory processes of homeostasis. Furthermore, macrophages are critically involved in initiating immunity, defending against injury and infection, and are also involved in immune resolution and wound healing. Heterogeneous populations of macrophage-type cells reside in many tissues and cause a variety of tissue-specific effects through direct or indirect interactions with other physiological systems, including the nervous and endocrine systems. It is vital to understand how macrophages respond to the unique environment of space to safeguard crew members with appropriate countermeasures for future missions in low Earth orbit and beyond. This review highlights current literature on macrophage responses to spaceflight and spaceflight analogs.
Collapse
Affiliation(s)
- Rocky An
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Cornell University, Department of Biological and Environmental Engineering and Sibley School of Mechanical and Aerospace Engineering, Ithaca, NY, 14853, USA
| | - Virginia Katherine Blackwell
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Massachusetts Institute of Technology, Department of Biology, Cambridge, MA, 02139, USA
| | - Bijan Harandi
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Tufts University, Department of Chemistry, Medford, MA, 02155, USA
| | - Alicia C Gibbons
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- University of California San Diego, Department of Cellular and Molecular Medicine, La Jolla, CA, 92093, USA
| | - Olivia Siu
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, 32114, USA
| | - Iris Irby
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Amy Rees
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Nadjet Cornejal
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Brooklyn College, Department of Natural and Behavioral Sciences, Brooklyn, NY, 11210, USA
| | - Kristina M Sattler
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- Ohio State University, Department of Physiology and Cell Biology, Columbus, OH, 43210, USA
| | - Tao Sheng
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- University of Pittsburgh, Department of Computer Science, Pittsburgh, PA, 15260, USA
| | - Nicholas C Syracuse
- NASA Ames Research Center, Space Life Sciences Training Program, Moffett Field, CA, 94035, USA
- North Carolina State University, Department of Molecular and Structural Biochemistry and Department of Biological Sciences, Raleigh, NC, 27695, USA
| | - David Loftus
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Sergio R Santa Maria
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Egle Cekanaviciute
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Sigrid S Reinsch
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
| | - Hami E Ray
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA
- ASRC Federal, Inc, Beltsville, MD, 20705, USA
| | - Amber M Paul
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, 32114, USA.
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA, 94035, USA.
- Blue Marble Space Institute of Science, Seattle, WA, 98104, USA.
| |
Collapse
|
24
|
Wu W, Ren J, Han M, Huang B. Influence of gut microbiome on metabolic diseases: a new perspective based on microgravity. J Diabetes Metab Disord 2024; 23:353-364. [PMID: 38932858 PMCID: PMC11196560 DOI: 10.1007/s40200-024-01394-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/28/2024] [Indexed: 06/28/2024]
Abstract
Purpose Microgravity, characterized by gravity levels of 10-3-10-6g, has been found to significantly impair various physiological systems in astronauts, including cardiovascular function, bone density, and metabolism. With the recent surge in human spaceflight, understanding the impact of microgravity on biological health has become paramount. Methods A comprehensive literature search was performed using the PubMed database to identify relevant publications pertaining to the interplay between gut microbiome, microgravity, space environment, and metabolic diseases. Results This comprehensive review primarily focuses on the progress made in investigating the gut microbiome and its association with metabolic diseases under microgravity conditions. Microgravity induces notable alterations in the composition, diversity, and functionality of the gut microbiome. These changes hold direct implications for metabolic disorders such as cardiovascular disease (CVD), bone metabolism disorders, energy metabolism dysregulation, liver dysfunction, and complications during pregnancy. Conclusion This novel perspective is crucial for preparing for deep space exploration and interstellar migration, where understanding the complex interplay between the gut microbiome and metabolic health becomes indispensable.
Collapse
Affiliation(s)
- Wanxin Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui China
| | - Junjie Ren
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, Anhui China
| | - Maozhen Han
- School of Life Sciences, Anhui Medical University, Hefei, 230032 Anhui China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui China
| |
Collapse
|
25
|
Suzuki K, Tsuruoka C, Morioka T, Seo H, Ogawa M, Kambe R, Imaoka T, Kakinuma S, Takahashi A. Combined effects of radiation and simulated microgravity on intestinal tumorigenesis in C3B6F1 Apc Min/+ mice. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:202-209. [PMID: 38670648 DOI: 10.1016/j.lssr.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Explorations of the Moon and Mars are planned as future manned space missions, during which humans will be exposed to both radiation and microgravity. We do not, however, know the health effects for such combined exposures. In a ground-based experiment, we evaluated the combined effects of radiation and simulated microgravity on tumorigenesis by performing X-irradiation and tail suspension in C3B6F1 ApcMin/+ mice, a well-established model for intestinal tumorigenesis. Mice were irradiated at 2 weeks of age and underwent tail suspension for 3 or 11 weeks using a special device that avoids damage to the tail. The tail suspension treatment significantly reduced the thymus weight after 3 weeks but not 11 weeks, suggesting a transient stress response. The combination of irradiation and tail suspension significantly increased the number of small intestinal tumors less than 2 mm in diameter as compared with either treatment alone. The combined treatment also increased the fraction of malignant tumors among all small intestinal tumors as compared with the radiation-only treatment. Thus, the C3B6F1 ApcMin/+ mouse is a useful model for assessing cancer risk in a simulated space environment, in which simulated microgravity accelerates tumor progression when combined with radiation exposure.
Collapse
Affiliation(s)
- Kenshi Suzuki
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hitomi Seo
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Mari Ogawa
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Ryosuke Kambe
- Gunma University Heavy Ion Medical Center, Gunma, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, Institute for Radiological Science (NIRS), National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| | | |
Collapse
|
26
|
Kimura Y, Nakai Y, Ino Y, Akiyama T, Moriyama K, Ohira T, Saito T, Inaba Y, Kumagai K, Ryo A, Hirano H. Identification of gravity-responsive serum proteins in spaceflight mice using a quantitative proteomic approach with data-independent acquisition mass spectrometry. Proteomics 2024; 24:e2300214. [PMID: 38475964 DOI: 10.1002/pmic.202300214] [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: 05/09/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Physical inactivity associated with gravity unloading, such as microgravity during spaceflight and hindlimb unloading (HU), can cause various physiological changes. In this study, we attempted to identify serum proteins whose levels fluctuated in response to gravity unloading. First, we quantitatively assessed changes in the serum proteome profiles of spaceflight mice using mass spectrometry with data-independent acquisition. The serum levels of several proteins involved in the responses to estrogen and glucocorticoid, blood vessel maturation, osteoblast differentiation, and ossification were changed by microgravity exposure. Furthermore, a collective evaluation of serum proteomic data from spaceflight and HU mice identified 30 serum proteins, including Mmp2, Igfbp2, Tnc, Cdh5, and Pmel, whose levels varied to a similar extent in both gravity unloading models. These changes in serum levels could be involved in the physiological changes induced by gravity unloading. A collective evaluation of serum, femur, and soleus muscle proteome data of spaceflight mice also showed 24 serum proteins, including Igfbp5, Igfbp3, and Postn, whose levels could be associated with biological changes induced by microgravity. This study examined serum proteome profiles in response to gravity unloading, and may help deepen our understanding of microgravity adaptation mechanisms during prolonged spaceflight missions.
Collapse
Affiliation(s)
- Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| |
Collapse
|
27
|
Zhou S, Li X, Liang F, Ji G, Lv K, Yuan Y, Zhao Y, Yan N, Zhang C, Cai S, Zhang S, Liu X, Song B, Qu L. Mitophagy Regulates the Circadian Rhythms by Degrading NR1D1 in Simulated Microgravity and Isolation Environments. Int J Mol Sci 2024; 25:4853. [PMID: 38732079 PMCID: PMC11084518 DOI: 10.3390/ijms25094853] [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: 03/07/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Long-term spaceflight is known to induce disruptions in circadian rhythms, which are driven by a central pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus, but the underlying molecular mechanisms remain unclear. Here, we developed a rat model that simulated microgravity and isolation environments through tail suspension and isolation (TSI). We found that the TSI environment imposed circadian disruptions to the core body temperature, heart rate, and locomotor-activity rhythms of rats, especially in the amplitude of these rhythms. In TSI model rats' SCNs, the core circadian gene NR1D1 showed higher protein but not mRNA levels along with decreased BMAL1 levels, which indicated that NR1D1 could be regulated through post-translational regulation. The autophagosome marker LC3 could directly bind to NR1D1 via the LC3-interacting region (LIR) motifs and induce the degradation of NR1D1 in a mitophagy-dependent manner. Defects in mitophagy led to the reversal of NR1D1 degradation, thereby suppressing the expression of BMAL1. Mitophagy deficiency and subsequent mitochondrial dysfunction were observed in the SCN of TSI models. Urolithin A (UA), a mitophagy activator, demonstrated an ability to enhance the amplitude of core body temperature, heart rate, and locomotor-activity rhythms by prompting mitophagy induction to degrade NR1D1. Cumulatively, our results demonstrate that mitophagy exerts circadian control by regulating NR1D1 degradation, revealing mitophagy as a potential target for long-term spaceflight as well as diseases with SCN circadian disruption.
Collapse
Affiliation(s)
- Sihai Zhou
- Department of Pathology and Forensics, Dalian Medical University, Dalian 116044, China;
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Xiaopeng Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Fengji Liang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Guohua Ji
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Ke Lv
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Yanhong Yuan
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Yujie Zhao
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Na Yan
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Chuanjie Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Shiou Cai
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Shuhui Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Xu Liu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| | - Bo Song
- Department of Pathology and Forensics, Dalian Medical University, Dalian 116044, China;
| | - Lina Qu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China; (X.L.); (F.L.); (G.J.); (K.L.); (Y.Y.); (Y.Z.); (N.Y.); (C.Z.); (S.C.); (X.L.)
| |
Collapse
|
28
|
Carnovali M, Zava S, Banfi G, Rizzo AM, Mariotti M. Vibration Rather than Microgravity Affects Bone Metabolism in Adult Zebrafish Scale Model. Cells 2024; 13:509. [PMID: 38534353 PMCID: PMC10969198 DOI: 10.3390/cells13060509] [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: 02/02/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Gravity and mechanical forces cause important alterations in the human skeletal system, as demonstrated by space flights. Innovative animal models like zebrafish embryos and medaka have been introduced to study bone response in ground-based microgravity simulators. We used, for the first time, adult zebrafish in simulated microgravity, with a random positioning machine (RPM) to study bone remodeling in the scales. To evaluate the effects of microgravity on bone remodeling in adult bone tissue, we exposed adult zebrafish to microgravity for 14 days using RPM and we evaluated bone remodeling on explanted scales. Our data highlight bone resorption in scales in simulated microgravity fish but also in the fish exposed, in normal gravity, to the vibrations produced by the RPM. The osteoclast activation in both rotating and non-rotating samples suggest that prolonged vibrations exposure leads to bone resorption in the scales tissue. Stress levels in these fish were normal, as demonstrated by blood cortisol quantification. In conclusion, vibrational mechanical stress induced bone resorption in adult fish scales. Moreover, adult fish as an animal model for microgravity studies remains controversial since fish usually live in weightless conditions because of the buoyant force from water and do not constantly need to support their bodies against gravity.
Collapse
Affiliation(s)
- Marta Carnovali
- IRCCS Ospedale Galeazzi Sant’Ambrogio, Via C. Belgioioso 173, 20161 Milan, Italy; (M.C.); (G.B.)
| | - Stefania Zava
- Department of Pharmacological and Biomedical Sciences “Rodolfo Paoletti”, University of Milan, Via D. Trentacoste 2, 20134 Milan, Italy; (S.Z.); (A.M.R.)
| | - Giuseppe Banfi
- IRCCS Ospedale Galeazzi Sant’Ambrogio, Via C. Belgioioso 173, 20161 Milan, Italy; (M.C.); (G.B.)
- School of Medicine, Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomedical Sciences “Rodolfo Paoletti”, University of Milan, Via D. Trentacoste 2, 20134 Milan, Italy; (S.Z.); (A.M.R.)
| | - Massimo Mariotti
- IRCCS Ospedale Galeazzi Sant’Ambrogio, Via C. Belgioioso 173, 20161 Milan, Italy; (M.C.); (G.B.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Commenda 10, 20122 Milan, Italy
| |
Collapse
|
29
|
Shama S, Ranade AV, Qaisar R, Khan NA, Tauseef I, Elmoselhi A, Siddiqui R. Enhancing microbial diversity as well as multi-organ health in hind-limb unloaded mice. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:62-71. [PMID: 38245349 DOI: 10.1016/j.lssr.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 01/22/2024]
Abstract
During space travel, the gut microbiota is changed which can lead to health-related issues. Previously, we utilized the hind-limb unloaded (HU) mouse, which is an established ground-based in-vivo model of microgravity and observed altered gut microbiota. In this study, we evaluated the beneficial effects of novel bacterial conditioned media in HU mice to understand if they can offset the effects of unloading in the HU mouse model. We aimed to explore the influence of bacterial conditioned media on diversity and quantity of intestinal microbes in HU mice, and investigated the microarchitecture of mice retinas and kidneys to evaluate the potential systemic effects of bacterial conditioned media in HU mice. Four-month-old, male C57/Bl6 mice were separated into groups: including the ground-based control group, the HU group mice fed with vehicle as placebo (HU-placebo mice), and the HU group fed with bacterial conditioned media (HU-CP mice) and kept under controlled environmental conditions for three weeks. Next, mice were sacrificed; gut dissections were conducted, and metagenomic analysis of bacterial species was performed via DNA extraction and 16S rRNA analysis. The results revealed an HU-induced reduction in intestinal microbial diversity, and an increase in pathogenic bacteria dominated by Firmicutes (45%). In contrast, supplementation with bacterial conditioned media for three weeks led to a significant increase in gut microbial diversity with noticeable changes in the OTUs abundance in the HU mice. Additionally, HU-induced muscle weakness and structural abnormalities in the retina and kidney were partially prevented with bacterial conditioned media. Moreover, a greater diversity of several bacteria in the HU-CP was observed including, Bacteriodota, Firmicutes, Proteobacteria, Actionobacteriota, Verrucomicorbiota, Cyanobacteria, Gemmatimonadota, Acidobacteriota, Chloroflexi, Myxococcota, and others. Prospective research involving molecular mechanistic studies are needed to comprehend the systemic effects of bacterial metabolites conditioned media on experimental animal models under chronic stress.
Collapse
Affiliation(s)
- Shama Shama
- Department of Clinical Sciences, College of Medicine, University of Sharjah, University City, Sharjah 27272, United Arab Emirates; Department of Microbiology, Hazara University, Mansehra 21300, Pakistan
| | - Anu V Ranade
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Naveed Ahmed Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey.
| | - Isfahan Tauseef
- Department of Microbiology, Hazara University, Mansehra 21300, Pakistan.
| | - Adel Elmoselhi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Ruqaiyyah Siddiqui
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey; College of Arts and Sciences, American University of Sharjah, University City, Sharjah 26666, United Arab Emirates
| |
Collapse
|
30
|
Kang L, Li Q, Jing Y, Ren F, Li E, Zeng X, Xu Y, Wang D, Wang Q, Sun G, Wei L, Diao Y. Auricularia auricula Anionic Polysaccharide Nanoparticles for Gastrointestinal Delivery of Pinus koraiensis Polyphenol Used in Bone Protection under Weightlessness. Molecules 2024; 29:245. [PMID: 38202827 PMCID: PMC10780251 DOI: 10.3390/molecules29010245] [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: 11/25/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Auricularia auricula polysaccharides used in Pinus koraiensis polyphenol encapsulation and delivery under weightlessness are rarely reported. In this study, an anionic polysaccharide fragment named AAP Iα with a molecular weight of 133.304 kDa was isolated and purified to construct a polyphenol encapsulation system. Nanoparticles named NPs-PP loaded with a rough surface for Pinus koraiensis polyphenol (PP) delivery were fabricated by AAP Iα and ε-poly-L-lysine (ε-PL). SEM and the DLS tracking method were used to observe continuous changes in AAP Iα, ε-PL and PP on the nanoparticles' rough surface assembly, as well as the dispersion and stability. Hydrophilic, monodisperse and highly negative charged nanoparticles can be formed at AAP Iα 0.8 mg/mL, ε-PL 20 μg/mL and PP 80 μg/mL. FT-IR was used to determine their electrostatic interactions. Release kinetic studies showed that nanoparticles had an ideal gastrointestinal delivery effect. NPs-PP loaded were assembled through electrostatic interactions between polyelectrolytes after hydrogen bonding formation in PP-AAP Iα and PP-ε-PL, respectively. Colon adhesion properties and PP delivery in vivo of nanoparticles showed that NPs-PP loaded had high adhesion efficiency to the colonic mucosa under simulated microgravity and could enhance PP bioavailability. These results suggest that AAP Iα can be used in PP encapsulation and delivery under microgravity in astronaut food additives.
Collapse
Affiliation(s)
- Li Kang
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
| | - Qiao Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China (E.L.)
| | - Yonghui Jing
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
| | - Feiyan Ren
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
| | - Erzhuo Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China (E.L.)
| | - Xiangyin Zeng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China (E.L.)
| | - Yier Xu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China (E.L.)
| | - Dongwei Wang
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
| | - Qiang Wang
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
| | - Guicai Sun
- The First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Lijun Wei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, China (E.L.)
| | - Yan Diao
- College of Life Science, China West Normal University, Nanchong 637009, China (Y.J.); (F.R.)
- Collaboration Innovation Center for Tissue Repair Material Engineering Technology, China West Normal University, Nanchong 637002, China
| |
Collapse
|
31
|
Ludtka C, Allen JB. The Effects of Simulated and Real Microgravity on Vascular Smooth Muscle Cells. GRAVITATIONAL AND SPACE RESEARCH : PUBLICATION OF THE AMERICAN SOCIETY FOR GRAVITATIONAL AND SPACE RESEARCH 2024; 12:46-59. [PMID: 38846256 PMCID: PMC11156189 DOI: 10.2478/gsr-2024-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
As considerations are being made for the limitations and safety of long-term human spaceflight, the vasculature is important given its connection to and impact on numerous organ systems. As a major constituent of blood vessels, vascular smooth muscle cells are of interest due to their influence over vascular tone and function. Additionally, vascular smooth muscle cells are responsive to pressure and flow changes. Therefore, alterations in these parameters under conditions of microgravity can be functionally disruptive. As such, here we review and discuss the existing literature that assesses the effects of microgravity, both actual and simulated, on smooth muscle cells. This includes the various methods for achieving or simulating microgravity, the animal models or cells used, and the various durations of microgravity assessed. We also discuss the various reported findings in the field, which include changes to cell proliferation, gene expression and phenotypic shifts, and renin-angiotensin-aldosterone system (RAAS), nitric oxide synthase (NOS), and Ca2+ signaling. Additionally, we briefly summarize the literature on smooth muscle tissue engineering in microgravity as well as considerations of radiation as another key component of spaceflight to contextualize spaceflight experiments, which by their nature include radiation exposure. Finally, we provide general recommendations based on the existing literature's focus and limitations.
Collapse
Affiliation(s)
- Christopher Ludtka
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Josephine B. Allen
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL
| |
Collapse
|
32
|
Kazmi S, Farajdokht F, Meynaghizadeh-Zargar R, Sadigh-Eteghad S, Pasokh A, Farzipour M, Farazi N, Hamblin MR, Mahmoudi J. Transcranial photobiomodulation mitigates learning and memory impairments induced by hindlimb unloading in a mouse model of microgravity exposure by suppression of oxidative stress and neuroinflammation signaling pathways. Brain Res 2023; 1821:148583. [PMID: 37717889 DOI: 10.1016/j.brainres.2023.148583] [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: 04/05/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Prolonged microgravity exposure causes cognitive impairment. Evidence shows that oxidative stress and neuroinflammation are involved in the causation. Here, we explore the effectiveness of transcranial near-infrared photobiomodulation (PBM) on cognitive deficits in a mouse model of simulated microgravity. 24 adult male C57BL/6 mice were assigned into three groups (8 in each); control, hindlimb unloading (HU), and HU + PBM groups. After surgery to fit the suspension fixing, the animals were housed either in HU cages or in their normal cage for 14 days. The mice in the HU + PBM group received PBM (810 nm laser, 10 Hz, 8 J/cm2) once per day for 14 days. Spatial learning and memory were assessed in the Lashley III maze and hippocampus tissue samples were collected to assess oxidative stress markers and protein expression of brain-derived neurotrophic factor (BDNF), nuclear factor erythroid 2-related factor 2 (Nrf2), Sirtuin 1 (Sirt1), and Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Behavioral testing showed that the PBM-treated animals had a shorter latency time to find the target and fewer errors than the HU group. PBM decreased hippocampal lipid peroxidation while increasing antioxidant defense systems (glutathione peroxidase, superoxide dismutase, and total antioxidant capacity) compared to HU mice. PBM increased protein expression of Sirt1, Nrf2, and BDNF while decreasing NF-κB compared to HU mice. Our findings suggested that the protective effect of PBM against HU-induced cognitive impairment involved the activation of the Sirt1/Nrf2 signaling pathway, up-regulation of BDNF, and reduction of neuroinflammation and oxidative stress in the hippocampus.
Collapse
Affiliation(s)
- Sareh Kazmi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pasokh
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Mohammad Farzipour
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narmin Farazi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Guo Q, Chen N, Patel K, Wan M, Zheng J, Cao X. Unloading-Induced Skeletal Interoception Alters Hypothalamic Signaling to Promote Bone Loss and Fat Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305042. [PMID: 37880864 PMCID: PMC10724445 DOI: 10.1002/advs.202305042] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/02/2023] [Indexed: 10/27/2023]
Abstract
Microgravity is the primary factor that affects human physiology in spaceflight, particularly bone loss and disturbances of the central nervous system. However, little is known about the cellular and molecular mechanisms of these effects. Here, it is reported that in mice hindlimb unloading stimulates expression of neuropeptide Y (NPY) and tyrosine hydroxylase (TH) in the hypothalamus, resulting in bone loss and altered fat metabolism. Enhanced expression of TH and NPY in the hypothalamus occurs downstream of a reduced prostaglandin E2 (PGE2)-mediated ascending interoceptive signaling of the skeletal interoception. Sympathetic antagonist propranolol or deletion of Adrb2 in osteocytes rescue bone loss in the unloading model. Moreover, depletion of TH+ sympathetic nerves or inhibition of norepinephrine release ameliorated bone resorption. Stereotactic inhibition of NPY expression in the hypothalamic neurons reduces the food intake with altered energy expenditure with a limited effect on bone, indicating hypothalamic neuroendocrine factor NPY in the facilitation of bone formation by sympathetic TH activity. These findings suggest that reduced PGE2-mediated interoceptive signaling in response to microgravity or unloading has impacts on the skeletal and central nervous systems that are reciprocally regulated.
Collapse
Affiliation(s)
- Qiaoyue Guo
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of EndocrinologyEndocrinology Research CenterXiangya Hospital of Central South UniversityChangshaHunan410008China
| | - Ningrong Chen
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Kalp Patel
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Mei Wan
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Junying Zheng
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Xu Cao
- Department of Orthopedic SurgeryJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
34
|
Zhou Y, Lv W, Peng X, Cheng Y, Tu Y, Song G, Luo Q. Simulated microgravity attenuates skin wound healing by inhibiting dermal fibroblast migration via F-actin/YAP signaling pathway. J Cell Physiol 2023; 238:2751-2764. [PMID: 37795566 DOI: 10.1002/jcp.31126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Skin and its cell components continuously subject to extrinsic and intrinsic mechanical forces and are mechanical sensitive. Disturbed mechanical homeostasis may lead to changes in skin functions. Gravity is the integral mechanical force on the earth, however, how gravity contributes to the maintenance of skin function and how microgravity in space affects the wound healing are poorly understood. Here, using microgravity analogs, we show that simulated microgravity (SMG) inhibits the healing of cutaneous wound and the accumulation of dermal fibroblasts in the wound bed. In vitro, SMG inhibits the migration of human foreskin fibroblast cells (HFF-1), and decreases the F-actin polymerization and YAP (yes-associated protein) activity. The SMG-inhibited migration can be recovered by activating YAP or F-actin polymerization using lysophosphatidic acid (LPA) or jasplakinolide (Jasp), suggesting the involvement of F-actin/YAP signaling pathway in this process. In SMG rats, LPA treatment improves the cutaneous healing with increased dermal fibroblasts in the wound bed. Together, our results demonstrate that SMG attenuates the cutaneous wound healing by inhibiting dermal fibroblast migration, and propose the crucial role of F-actin/YAP mechano-transduction in the maintenance of skin homeostasis under normal gravity, and YAP as a possible therapeutic target for the skin care of astronauts in space.
Collapse
Affiliation(s)
- Yuhao Zhou
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Wenjun Lv
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Xiufen Peng
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Yansiwei Cheng
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Yun Tu
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, College of Bioengineering, Ministry of Education, Chongqing University, Chongqing, China
| |
Collapse
|
35
|
Andrade MR, Azeez TA, Montgomery MM, Caldwell JT, Park H, Kwok AT, Borg AM, Narayanan SA, Willey JS, Delp MD, La Favor JD. Neurovascular dysfunction associated with erectile dysfunction persists after long-term recovery from simulations of weightlessness and deep space irradiation. FASEB J 2023; 37:e23246. [PMID: 37990646 DOI: 10.1096/fj.202300506rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 11/23/2023]
Abstract
There has been growing interest within the space industry for long-duration manned expeditions to the Moon and Mars. During deep space missions, astronauts are exposed to high levels of galactic cosmic radiation (GCR) and microgravity which are associated with increased risk of oxidative stress and endothelial dysfunction. Oxidative stress and endothelial dysfunction are causative factors in the pathogenesis of erectile dysfunction, although the effects of spaceflight on erectile function have been unexplored. Therefore, the purpose of this study was to investigate the effects of simulated spaceflight and long-term recovery on tissues critical for erectile function, the distal internal pudendal artery (dIPA), and the corpus cavernosum (CC). Eighty-six adult male Fisher-344 rats were randomized into six groups and exposed to 4-weeks of hindlimb unloading (HLU) or weight-bearing control, and sham (0Gy), 0.75 Gy, or 1.5 Gy of simulated GCR at the ground-based GCR simulator at the NASA Space Radiation Laboratory. Following a 12-13-month recovery, ex vivo physiological analysis of the dIPA and CC tissue segments revealed differential impacts of HLU and GCR on endothelium-dependent and -independent relaxation that was tissue type specific. GCR impaired non-adrenergic non-cholinergic (NANC) nerve-mediated relaxation in the dIPA and CC, while follow-up experiments of the CC showed restoration of NANC-mediated relaxation of GCR tissues following acute incubation with the antioxidants mito-TEMPO and TEMPOL, as well as inhibitors of xanthine oxidase and arginase. These findings indicate that simulated spaceflight exerts a long-term impairment of neurovascular erectile function, which exposes a new health risk to consider with deep space exploration.
Collapse
Affiliation(s)
- Manuella R Andrade
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Tooyib A Azeez
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - McLane M Montgomery
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Jacob T Caldwell
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Hyerim Park
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Andy T Kwok
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Alexander M Borg
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - S Anand Narayanan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael D Delp
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Justin D La Favor
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
36
|
Rosa-Caldwell ME, Mortreux M, Wadhwa A, Kaiser UB, Sung DM, Bouxsein ML, Rutkove SB. Sex differences in muscle health in simulated micro- and partial-gravity environments in rats. SPORTS MEDICINE AND HEALTH SCIENCE 2023; 5:319-328. [PMID: 38314043 PMCID: PMC10831389 DOI: 10.1016/j.smhs.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/22/2023] [Accepted: 09/06/2023] [Indexed: 02/06/2024] Open
Abstract
Skeletal muscle size and strength are important for overall health for astronauts. However, how male and female muscle may respond differently to micro- and partial-gravity environments is not fully understood. The purpose of this study was to determine how biological sex and sex steroid hormones influence the progression of muscle atrophy after long term exposure to micro and partial gravity environments in male and female rats. Male and female Fisher rats (n = 120) underwent either castration/ovariectomy or sham surgeries. After two weeks recovery, animals were divided into microgravity (0g), partial-gravity (40% of weight bearing, 0.4g), or full weight bearing (1g) interventions for 28 days. Measurements of muscle size and strength were evaluated prior to and after interventions. At 0g, females lost more dorsiflexion strength, plantar flexion strength, and other metrics of muscle size compared to males; castration/ovariectomy did not influence these differences. Additionally, at 0.4g, females lost more dorsiflexion strength, plantar flexion strength, and other metrics of muscle strength compared to males; castration/ovariectomy did not influence these differences. Females have greater musculoskeletal aberrations during exposure to both microgravity and partial-gravity environments; these differences are not dependent on the presence of sex steroid hormones. Correspondingly, additional interventions may be necessary to mitigate musculoskeletal loss in female astronauts to protect occupational and overall health.
Collapse
Affiliation(s)
- Megan E. Rosa-Caldwell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Marie Mortreux
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Department of Nutrition and Food Sciences, University of Rhode Island, Kingston, RI, 02881, USA
| | - Anna Wadhwa
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Ursula B. Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Womenʼs Hospital and Harvard Medical School, Boston, MA, 02215, USA
| | - Dong-Min Sung
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Mary L. Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Seward B. Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| |
Collapse
|
37
|
Fonte C, Jacob P, Vanet A, Ghislin S, Frippiat JP. Hindlimb unloading, a physiological model of microgravity, modifies the murine bone marrow IgM repertoire in a similar manner as aging but less strongly. Immun Ageing 2023; 20:64. [PMID: 37986079 PMCID: PMC10659048 DOI: 10.1186/s12979-023-00393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/12/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND The spaceflight environment is an extreme environment that affects the immune system of approximately 50% of astronauts. With planned long-duration missions, such as the deployment of the Lunar Gateway and possible interplanetary missions, it is mandatory to determine how all components of the immune system are affected, which will allow the establishment of countermeasures to preserve astronaut health. However, despite being an important component of the immune system, antibody-mediated humoral immunity has rarely been investigated in the context of the effects of the space environment. It has previously been demonstrated that 30 days aboard the BION-M1 satellite and 21 days of hindlimb unloading (HU), a model classically used to mimic the effects of microgravity, decrease murine B lymphopoiesis. Furthermore, modifications in B lymphopoiesis reported in young mice subjected to 21 days of HU were shown to be similar to those observed in aged mice (18-22 months). Since the primary antibody repertoire composed of IgM is created by V(D) J recombination during B lymphopoiesis, the objective of this study was to assess the degree of similarity between changes in the bone marrow IgM repertoire and in the V(D)J recombination process in 2.5-month-old mice subjected to 21 days of HU and aged (18 months) mice. RESULTS We found that in 21 days, HU induced changes in the IgM repertoire that were approximately 3-fold less than those in aged mice, which is a rapid effect. Bone remodeling and epigenetics likely mediate these changes. Indeed, we previously demonstrated a significant decrease in tibial morphometric parameters from day 6 of HU and a progressive reduction in these parameters until day 21 of HU, and it has been shown that age and microgravity induce epigenetic changes. CONCLUSION These data reveal novel immune changes that are akin to advanced aging and underline the importance of studying the effects of spaceflight on antibody-mediated humoral immunity.
Collapse
Affiliation(s)
- Coralie Fonte
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Pauline Jacob
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Anne Vanet
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Stéphanie Ghislin
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Jean-Pol Frippiat
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France.
| |
Collapse
|
38
|
Ino Y, Ohira T, Kumagai K, Nakai Y, Akiyama T, Moriyama K, Takeda Y, Saito T, Ryo A, Inaba Y, Hirano H, Kimura Y. Identification of mouse soleus muscle proteins altered in response to changes in gravity loading. Sci Rep 2023; 13:15768. [PMID: 37737267 PMCID: PMC10517164 DOI: 10.1038/s41598-023-42875-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
Gravity-dependent physical processes strongly affect the ability of elderly people to maintain musculoskeletal health by reducing muscle atrophy and increasing bone mineral density, thereby increasing quality of life. A need therefore exists to identify molecules in the musculoskeletal system that are responsive to gravitational loading and to establish an objective indicator for the maintenance of healthy musculoskeletal systems. Here, we performed an integrated assessment of the results of soleus muscle proteomic analyses in three model mouse experiments under different gravity environments (hypergravity, hindlimb unloading, and spaceflight). Myl6b, Gpd1, Fbp2, Pvalb, and Actn3 were shown to be gravity-responsive muscle proteins, and alterations in the levels of these proteins indicated changes in muscle fiber type to slow-twitch type due to gravity loading. In addition, immunoblotting and enzyme-linked immunosorbent assays revealed that Pvalb levels in the sera of hindlimb-unloaded mice and osteoporosis patients were higher than in control subjects, suggesting that Pvalb levels might be useful to objectively evaluate soleus muscle atrophy and bone loss.
Collapse
Affiliation(s)
- Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan.
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Ohno-Higashi 377-2, Osaka-Sayama, Osaka, Japan.
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yuriko Takeda
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | | | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
39
|
Stern C, Yücel YH, Zu Eulenburg P, Pavy-Le Traon A, Petersen LG. Eye-brain axis in microgravity and its implications for Spaceflight Associated Neuro-ocular Syndrome. NPJ Microgravity 2023; 9:56. [PMID: 37474624 PMCID: PMC10359255 DOI: 10.1038/s41526-023-00300-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/02/2023] [Indexed: 07/22/2023] Open
Abstract
Long-duration human spaceflight can lead to changes in both the eye and the brain, which have been referred to as Spaceflight Associated Neuro-ocular Syndrome (SANS). These changes may manifest as a constellation of symptoms, which can include optic disc edema, optic nerve sheath distension, choroidal folds, globe flattening, hyperopic shift, and cotton wool spots. Although the underpinning mechanisms for SANS are not yet known, contributors may include intracranial interstitial fluid accumulation following microgravity induced headward fluid shift. Development and validation of SANS countermeasures contribute to our understanding of etiology and accelerate new technology including exercise modalities, Lower Body Negative Pressure suits, venous thigh cuffs, and Impedance Threshold Devices. However, significant knowledge gaps remain including biomarkers, a full set of countermeasures and/or treatment regimes, and finally reliable ground based analogs to accelerate the research. This review from the European Space Agency SANS expert group summarizes past research and current knowledge on SANS, potential countermeasures, and key knowledge gaps, to further our understanding, prevention, and treatment of SANS both during human spaceflight and future extraterrestrial surface exploration.
Collapse
Affiliation(s)
- Claudia Stern
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany.
- ISS Operations and Astronauts Group, European Astronaut Centre, European Space Agency (ESA), Cologne, Germany.
| | - Yeni H Yücel
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Department of Ophthalmology & Vision Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Physics, Faculty of Science, Toronto Metropolitan University, Toronto, ON, Canada
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Peter Zu Eulenburg
- Institute for Neuroradiology & German Center for Vertigo and Balance Disorders, Ludwig-Maximilians-University, Munich, Germany
| | - Anne Pavy-Le Traon
- Department of Neurology, University Hospital of Toulouse, Toulouse, France
- MEDES, Institute for Space Physiology and Medicine, Toulouse, France
- UMR INSERM U1297, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
| | - Lonnie Grove Petersen
- Department of Aeronautics and Astronautics, Massachusetts Institute of Technology, Cambridge, USA
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Naumova AA, Oleynik EA, Khramtsova AV, Nikolaeva SD, Chernigovskaya EV, Glazova MV. Short-term hindlimb unloading negatively affects dopaminergic transmission in the nigrostriatal system of mice. Dev Neurobiol 2023; 83:205-218. [PMID: 37489016 DOI: 10.1002/dneu.22924] [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: 02/14/2023] [Revised: 07/04/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
The nigrostriatal system composed of the dorsal striatum and the substantia nigra (SN) is highly involved in the control of motor behavior. Various extremal and pathological conditions as well as social isolation (SI) may cause an impairment of locomotor function; however, corresponding alterations in the nigrostriatal dopaminergic pathway are far from full understanding. Here, we analyzed the effect of 3-day hindlimb unloading (HU) and SI on the key players of dopamine transmission in the nigrostriatal system of CD1 mice. Three groups of mice were analyzed: group-housed (GH), SI, and HU animals. Our data showed a significant decrease in the expression and phosphorylation of tyrosine hydroxylase (TH) in the SN and dorsal striatum of HU mice that suggested attenuation of dopamine synthesis in response to HU. In the dorsal striatum of HU mice, the downregulation of TH expression was also observed indicating the effect of unloading; however, TH phosphorylation at Ser40 was mainly affected by SI pointing on an impact of isolation too. Expression of dopamine receptors D1 in the dorsal striatum of HU mice was increased suggesting a compensatory response, but the activity of downstream signaling pathways involving protein kinase A and cAMP response element-binding protein was inhibited. At the same time, SI alone did not affect expression of DA receptors and activity of downstream signaling in the dorsal striatum. Obtained data let us to conclude that HU was the main factor which impaired dopamine transmission in the nigrostriatal system but SI made some contribution to its negative effects.
Collapse
Affiliation(s)
- Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Ekaterina A Oleynik
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Anna V Khramtsova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Svetlana D Nikolaeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Elena V Chernigovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
41
|
Huang Y, Dou Y, Yang B, He B, Zhang X, Zhang K, Yang X. Nicotinamide mononucleotide supplementation mitigates osteopenia induced by modeled microgravity in rats. Cell Stress Chaperones 2023; 28:385-394. [PMID: 37195399 PMCID: PMC10352228 DOI: 10.1007/s12192-023-01356-7] [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: 04/30/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023] Open
Abstract
Exposure to weightlessness causes severe osteopenia, resulting in raised fracture risk. The current study aimed to investigate whether nicotinamide mononucleotide (NMN) supplementation protected against the osteopenia in hindlimb unloading (HLU) rats in vivo and modeled microgravity-induced osteoblastic dysfunction in vitro. The 3-mo-old rats were exposed to HLU and intragastrically administered NMN every 3 days (500 mg/kg body weight) for 4 weeks. NMN supplementation mitigated HLU-induced bone loss, evidenced by greater bone mass and biomechanical properties and better trabecular bone structure. NMN supplementation mitigated HLU-induced oxidative stress, evidenced by greater levels of nicotinamide adenine dinucleotide and activities of superoxide dismutase 2 and lesser malondialdehyde levels. Modeled microgravity stimulation using rotary wall vessel bioreactor in MC3T3-E1 cells inhibited osteoblast differentiation, which was reversed by NMN treatment. Furthermore, NMN treatment mitigated microgravity-induced mitochondrial impairments, evidenced by lesser reactive oxygen species generation and greater adenosine triphosphate production, mtDNA copy number, and activities of superoxide dismutase 2 and Complex I and II. Additionally, NMN promoted activation of AMP-activated protein kinase (AMPK), evidenced by greater AMPKα phosphorylation. Our research suggested that NMN supplementation attenuated osteoblastic mitochondrial impairment and mitigated osteopenia induced by modeled microgravity.
Collapse
Affiliation(s)
- Yunfei Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Yusheng Dou
- Department of Should and Elbow Joint, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bo Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Baorong He
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xuefang Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Ke Zhang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China
| | - Xiaobin Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, No.99 Yanxiang Road, Xi'an, 710054, Shaanxi Province, China.
| |
Collapse
|
42
|
Markina E, Andreeva E, Buravkova L. Stromal Lineage Precursors from Rodent Femur and Tibia Bone Marrows after Hindlimb Unloading: Functional Ex Vivo Analysis. Int J Mol Sci 2023; 24:ijms24108594. [PMID: 37239936 DOI: 10.3390/ijms24108594] [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: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Rodent hindlimb unloading (HU) model was developed to elucidate responses/mechanisms of adverse consequences of space weightlessness. Multipotent mesenchymal stromal cells (MMSCs) were isolated from rat femur and tibia bone marrows and examined ex vivo after 2 weeks of HU and subsequent 2 weeks of restoration of load (HU + RL). In both bones, decrease of fibroblast colony forming units (CFU-f) after HU with restoration after HU + RL detected. In CFU-f and MMSCs, levels of spontaneous/induced osteocommitment were similar. MMSCs from tibia initially had greater spontaneous mineralization of extracellular matrix but were less sensitive to osteoinduction. There was no recovery of initial levels of mineralization in MMSCs from both bones during HU + RL. After HU, most bone-related genes were downregulated in tibia or femur MMSCs. After HU + RL, the initial level of transcription was restored in femur, while downregulation persisted in tibia MMSCs. Therefore, HU provoked a decrease of osteogenic activity of BM stromal precursors at transcriptomic and functional levels. Despite unidirectionality of changes, the negative effects of HU were more pronounced in stromal precursors from distal limb-tibia. These observations appear to be on demand for elucidation of mechanisms of skeletal disorders in astronauts in prospect of long-term space missions.
Collapse
Affiliation(s)
- Elena Markina
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Elena Andreeva
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Ludmila Buravkova
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| |
Collapse
|
43
|
Buettmann EG, DeNapoli RC, Abraham LB, Denisco JA, Lorenz MR, Friedman MA, Donahue HJ. Reambulation following hindlimb unloading attenuates disuse-induced changes in murine fracture healing. Bone 2023; 172:116748. [PMID: 37001629 DOI: 10.1016/j.bone.2023.116748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Patients with bone and muscle loss from prolonged disuse have higher risk of falls and subsequent fragility fractures. In addition, fracture patients with continued disuse and/or delayed physical rehabilitation have worse clinical outcomes compared to individuals with immediate weight-bearing activity following diaphyseal fracture. However, the effects of prior disuse followed by physical reambulation on fracture healing cellular processes and adjacent bone and skeletal muscle recovery post-injury remains poorly defined. To bridge this knowledge gap and inform future treatment and rehabilitation strategies for fractures, a preclinical model of fracture healing with a history of prior unloading with and without reambulation was employed. First, skeletally mature male and female C57BL/6J mice (18 weeks) underwent hindlimb unloading by tail suspension (HLU) for 3 weeks to induce significant bone and muscle loss modeling enhanced bone fragility. Next, mice had their right femur fractured by open surgical dissection (stabilized with 24-gauge pin). The, mice were randomly assigned to continued HLU or allowed normal weight-bearing reambulation (HLU + R). Mice given normal cage activity throughout the experiment served as healthy age-matched controls. All mice were sacrificed 4-days (DPF4) or 14-days (DPF14) following fracture to assess healing and uninjured hindlimb musculoskeletal properties (6-10 mice per treatment/biological sex). We found that continued disuse following fracture lead to severely diminished uninjured hindlimb skeletal muscle mass (gastrocnemius and soleus) and femoral bone volume adjacent to the fracture site compared to healthy age-matched controls across mouse sexes. Furthermore, HLU led to significantly decreased periosteal expansion (DPF4) and osteochondral tissue formation by DPF14, and trends in increased osteoclastogenesis (DPF14) and decreased woven bone vascular area (DPF14). In contrast, immediate reambulation for 2 weeks after fracture, even following a period of prolonged disuse, was able to increase hindlimb skeletal tissue mass and increase osteochondral tissue formation, albeit not to healthy control levels, in both mouse sexes. Furthermore, reambulation attenuated osteoclast formation seen in woven bone tissue undergoing disuse. Our results suggest that weight-bearing skeletal loading in both sexes immediately following fracture may improve callus healing and prevent further fall risk by stimulating skeletal muscle anabolism and decreasing callus resorption compared to minimal or delayed rehabilitation regimens.
Collapse
Affiliation(s)
- Evan G Buettmann
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Rachel C DeNapoli
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Lovell B Abraham
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Joe A Denisco
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Madelyn R Lorenz
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Michael A Friedman
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America
| | - Henry J Donahue
- Virginia Commonwealth University, Biomedical Engineering, Richmond, VA, United States of America.
| |
Collapse
|
44
|
Corydon TJ, Schulz H, Richter P, Strauch SM, Böhmer M, Ricciardi DA, Wehland M, Krüger M, Erzinger GS, Lebert M, Infanger M, Wise PM, Grimm D. Current Knowledge about the Impact of Microgravity on Gene Regulation. Cells 2023; 12:cells12071043. [PMID: 37048115 PMCID: PMC10093652 DOI: 10.3390/cells12071043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Microgravity (µg) has a massive impact on the health of space explorers. Microgravity changes the proliferation, differentiation, and growth of cells. As crewed spaceflights into deep space are being planned along with the commercialization of space travelling, researchers have focused on gene regulation in cells and organisms exposed to real (r-) and simulated (s-) µg. In particular, cancer and metastasis research benefits from the findings obtained under µg conditions. Gene regulation is a key factor in a cell or an organism’s ability to sustain life and respond to environmental changes. It is a universal process to control the amount, location, and timing in which genes are expressed. In this review, we provide an overview of µg-induced changes in the numerous mechanisms involved in gene regulation, including regulatory proteins, microRNAs, and the chemical modification of DNA. In particular, we discuss the current knowledge about the impact of microgravity on gene regulation in different types of bacteria, protists, fungi, animals, humans, and cells with a focus on the brain, eye, endothelium, immune system, cartilage, muscle, bone, and various cancers as well as recent findings in plants. Importantly, the obtained data clearly imply that µg experiments can support translational medicine on Earth.
Collapse
Affiliation(s)
- Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus, Denmark
- Correspondence: ; Tel.: +45-28-992-179
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Peter Richter
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, 91058 Erlangen, Germany
| | - Sebastian M. Strauch
- Postgraduate Program in Health and Environment, University of Joinville Region, Joinville 89219-710, SC, Brazil
| | - Maik Böhmer
- Institute for Molecular Biosciences, Johann Wolfgang Goethe Universität, 60438 Frankfurt am Main, Germany
| | - Dario A. Ricciardi
- Institute for Molecular Biosciences, Johann Wolfgang Goethe Universität, 60438 Frankfurt am Main, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Gilmar S. Erzinger
- Postgraduate Program in Health and Environment, University of Joinville Region, Joinville 89219-710, SC, Brazil
| | - Michael Lebert
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, 91058 Erlangen, Germany
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Microgravity and Translational Regenerative Medicine, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
45
|
Sanders LM, Scott RT, Yang JH, Qutub AA, Garcia Martin H, Berrios DC, Hastings JJA, Rask J, Mackintosh G, Hoarfrost AL, Chalk S, Kalantari J, Khezeli K, Antonsen EL, Babdor J, Barker R, Baranzini SE, Beheshti A, Delgado-Aparicio GM, Glicksberg BS, Greene CS, Haendel M, Hamid AA, Heller P, Jamieson D, Jarvis KJ, Komarova SV, Komorowski M, Kothiyal P, Mahabal A, Manor U, Mason CE, Matar M, Mias GI, Miller J, Myers JG, Nelson C, Oribello J, Park SM, Parsons-Wingerter P, Prabhu RK, Reynolds RJ, Saravia-Butler A, Saria S, Sawyer A, Singh NK, Snyder M, Soboczenski F, Soman K, Theriot CA, Van Valen D, Venkateswaran K, Warren L, Worthey L, Zitnik M, Costes SV. Biological research and self-driving labs in deep space supported by artificial intelligence. NAT MACH INTELL 2023. [DOI: 10.1038/s42256-023-00618-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
46
|
Hélissen O, Kermorgant M, Déjean S, Mercadie A, Le Gonidec S, Zahreddine R, Calise D, Nasr N, Galès C, Arvanitis DN, Pavy-Le Traon A. Autonomic Nervous System Adaptation and Circadian Rhythm Disturbances of the Cardiovascular System in a Ground-Based Murine Model of Spaceflight. Life (Basel) 2023; 13:life13030844. [PMID: 36983999 PMCID: PMC10057816 DOI: 10.3390/life13030844] [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: 02/06/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Whether in real or simulated microgravity, Humans or animals, the kinetics of cardiovascular adaptation and its regulation by the autonomic nervous system (ANS) remain controversial. In this study, we used hindlimb unloading (HU) in 10 conscious mice. Blood pressure (BP), heart rate (HR), temperature, and locomotor activity were continuously monitored with radio-telemetry, during 3 days of control, 5 days of HU, and 2 days of recovery. Six additional mice were used to assess core temperature. ANS activity was indirectly determined by analyzing both heart rate variability (HRV) and baroreflex sensitivity (BRS). Our study showed that HU induced an initial bradycardia, accompanied by an increase in vagal activity markers of HRV and BRS, together with a decrease in water intake, indicating the early adaptation to fluid redistribution. During HU, BRS was reduced; temperature and BP circadian rhythms were altered, showing a loss in day/night differences, a decrease in cycle amplitude, a drop in core body temperature, and an increase in day BP suggestive of a rise in sympathetic activity. Reloading induced resting tachycardia and a decrease in BP, vagal activity, and BRS. In addition to cardiovascular deconditioning, HU induces disruption in day/night rhythmicity of locomotor activity, temperature, and BP.
Collapse
Affiliation(s)
- Ophélie Hélissen
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
| | - Marc Kermorgant
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
- Neurology Department, University Hospital of Toulouse, 31400 Toulouse, France
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse, UMR5219, CNRS, Université de Toulouse, UT3, 31062 Toulouse, France
| | - Aurélie Mercadie
- Institut de Mathématiques de Toulouse, UMR5219, CNRS, Université de Toulouse, UT3, 31062 Toulouse, France
| | - Sophie Le Gonidec
- CREFRE-Anexplo, Services Phénotypage et Microchirurgie, UMS006, INSERM, Université de Toulouse, UT3, ENVT, 31062 Toulouse, France
| | - Rana Zahreddine
- CREFRE-Anexplo, Services Phénotypage et Microchirurgie, UMS006, INSERM, Université de Toulouse, UT3, ENVT, 31062 Toulouse, France
| | - Denis Calise
- CREFRE-Anexplo, Services Phénotypage et Microchirurgie, UMS006, INSERM, Université de Toulouse, UT3, ENVT, 31062 Toulouse, France
| | - Nathalie Nasr
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
| | - Céline Galès
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
| | - Dina N Arvanitis
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
| | - Anne Pavy-Le Traon
- Institute of Cardiovascular and Metabolic Diseases, UMR1297, INSERM, University Hospital of Toulouse, 31400 Toulouse, France
- Neurology Department, University Hospital of Toulouse, 31400 Toulouse, France
| |
Collapse
|
47
|
Ranade AV, Khan AA, Gul MT, Jose J, Ramachandran G, Qaisar R, Karim A, Ahmad F, Abdel-Rahman WM. Pharmacological inhibition of endoplasmic reticulum stress mitigates testicular pathology in a mouse model of simulated microgravity. ACTA ASTRONAUTICA 2023; 204:466-476. [DOI: 10.1016/j.actaastro.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
|
48
|
Chen F, Jiang N, Zhang YW, Xie MZ, Liu XM. Protective effect of Gastrodia elata blume ameliorates simulated weightlessness-induced cognitive impairment in mice. LIFE SCIENCES IN SPACE RESEARCH 2023; 36:1-7. [PMID: 36682818 DOI: 10.1016/j.lssr.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/19/2022] [Accepted: 10/02/2022] [Indexed: 06/17/2023]
Abstract
During the long-term orbital flight, exposure to microgravity negatively affects the astronauts' development of cognition, characterized by learning and memory decline. Gastrodia elata Blume (GEB) has a significant protective effect on cognitive impairment and has been used in Asia for centuries as a functional product. A previous study demonstrated that GEB could improve memory loss in mice caused by circadian rhythm disorders. However, the effects of GEB on cognitive dysfunction caused by weightless environments have not been investigated. In this study, mice received daily treatment with GEB (0.5, 1 g·kg-1d-1, i.g) and Huperzine A(Hup, 0.1 mg·kg-1d-1, i.g) orally until the end of the behavioral test (New object recognition test (NORT). Malondialdehyde (MDA) and nitric oxide (NO) levels were detected by kits, and expression of brain-derived neurotrophic factor (BDNF), protein kinase B (AKT), phosphorylated Akt (P-AKT), synaptophysin (SYN) and postsynaptic density 95(PSD95) in hippocampus were detected by western blotting. The results show that administration of GEB (0.5, 1 g·kg-1d-1, i.g) and Hup (0.1 mg·kg-1d-1, i.g) remarkably reverse HLS-induced learning and behavioral memory disorders, which were associated with significant changes in MDA and NO levels. Additionally, the protein expressions of BDNF, P-AKT/AKT, SYN, and PSD95 were significantly increased in the hippocampus. In summary, our findings will improve the reference for developing GEB as a functional product that improves memory decline.
Collapse
Affiliation(s)
- Fang Chen
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Hunan University of Chinese Medicine, Hunan 410000, China
| | - Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yi Wen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Meng Zhou Xie
- Hunan University of Chinese Medicine, Hunan 410000, China.
| | - Xin Min Liu
- Institute of New Drug Technology, Ningbo University, Ningbo 315000, China; Hunan University of Chinese Medicine, Hunan 410000, China.
| |
Collapse
|
49
|
Combined space stressors induce independent behavioral deficits predicted by early peripheral blood monocytes. Sci Rep 2023; 13:1749. [PMID: 36720960 PMCID: PMC9889764 DOI: 10.1038/s41598-023-28508-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/19/2023] [Indexed: 02/02/2023] Open
Abstract
Interplanetary space travel poses many hazards to the human body. To protect astronaut health and performance on critical missions, there is first a need to understand the effects of deep space hazards, including ionizing radiation, confinement, and altered gravity. Previous studies of rodents exposed to a single such stressor document significant deficits, but our study is the first to investigate possible cumulative and synergistic impacts of simultaneous ionizing radiation, confinement, and altered gravity on behavior and cognition. Our cohort was divided between 6-month-old female and male mice in group, social isolation, or hindlimb unloading housing, exposed to 0 or 50 cGy of 5 ion simplified simulated galactic cosmic radiation (GCRsim). We report interactions and independent effects of GCRsim exposure and housing conditions on behavioral and cognitive performance. Exposure to GCRsim drove changes in immune cell populations in peripheral blood collected early after irradiation, while housing conditions drove changes in blood collected at a later point. Female mice were largely resilient to deficits observed in male mice. Finally, we used principal component analysis to represent total deficits as principal component scores, which were predicted by general linear models using GCR exposure, housing condition, and early blood biomarkers.
Collapse
|
50
|
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
|