1
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
2
|
Mei T, Chen Y, Gao Y, Zhao H, Lyu X, Lin J, Niu T, Han H, Tong Z. Formaldehyde initiates memory and motor impairments under weightlessness condition. NPJ Microgravity 2024; 10:100. [PMID: 39468074 PMCID: PMC11519943 DOI: 10.1038/s41526-024-00441-0] [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: 10/27/2023] [Accepted: 10/21/2024] [Indexed: 10/30/2024] Open
Abstract
During space flight, prolonged weightlessness stress exerts a range of detrimental impacts on the physiology and psychology of astronauts. These manifestations encompass depressive symptoms, anxiety, and impairments in both short-term memory and motor functions, albeit the precise underlying mechanisms remain elusive. Recent studies have revealed that hindlimb unloading (HU) animal models, which simulate space weightlessness, exhibited a disorder in memory and motor function associated with endogenous formaldehyde (FA) accumulation in the hippocampus and cerebellum, disruption of brain extracellular space (ECS), and blockage of interstitial fluid (ISF) drainage. Notably, the impairment of the blood-brain barrier (BBB) caused by space weightlessness elicits the infiltration of albumin and hemoglobin from the blood vessels into the brain ECS. However, excessive FA has the potential to form cross-links between these two proteins and amyloid-beta (Aβ), thereby obstructing ECS and inducing neuron death. Moreover, FA can inhibit N-methyl-D-aspartate (NMDA) currents by crosslinking NR1 and NR2B subunits, thus impairing memory. Additionally, FA has the ability to modulate the levels of certain microRNAs (miRNAs) such as miRNA-29b, which can affect the expression of aquaporin-4 (AQP4) so as to regulate ECS structure and ISF drainage. Especially, the accumulation of FA may inactivate the ataxia telangiectasia-mutated (ATM) protein kinase by forming cross-linking, a process that is associated with ataxia. Hence, this review presents that weightlessness stress-derived FA may potentially serve as a crucial catalyst in the deterioration of memory and motor abilities in the context of microgravity.
Collapse
Affiliation(s)
- Tianhao Mei
- Beijing Geriatric Hospital, Beijing, China
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Chen
- Beijing Geriatric Hospital, Beijing, China
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yajuan Gao
- Department of Radiology, Peking University Third Hospital, Beijing, China. Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
- NMPA key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Hang Zhao
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xingzhou Lyu
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Lin
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianye Niu
- Shenzhen Bay Laboratory, Shenzhen, China.
- University of Science and Technology of China, Anhui, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China. Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
- NMPA key Laboratory for Evaluation of Medical Imaging Equipment and Technique, Beijing, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China.
| | - Zhiqian Tong
- Beijing Geriatric Hospital, Beijing, China.
- Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
3
|
Oommen AM, Stafford P, Joshi L. Profiling muscle transcriptome in mice exposed to microgravity using gene set enrichment analysis. NPJ Microgravity 2024; 10:94. [PMID: 39367013 PMCID: PMC11452717 DOI: 10.1038/s41526-024-00434-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: 02/09/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Space exploration's advancement toward long-duration missions prompts intensified research on physiological effects. Despite adaptive physiological stability in some variables, persistent changes affect genome integrity, immune response, and cognitive function. Our study, utilizing multi-omics data from GeneLab, provides crucial insights investigating muscle atrophy during space mission. Leveraging NASA GeneLab's data resources, we apply systems biology-based analyses, facilitating comprehensive understanding and enabling meta-analysis. Through transcriptomics, we establish a reference profile of biological processes underlying muscle atrophy, crucial for intervention development. We emphasize the often-overlooked role of glycosylation in muscle atrophy. Our research sheds light on fundamental molecular mechanisms, bridging gaps between space research and terrestrial conditions. This study underscores the importance of interdisciplinary collaboration and data-sharing initiatives like GeneLab in advancing space medicine research.
Collapse
Affiliation(s)
- Anup Mammen Oommen
- Advanced Glycoscience Research Cluster (AGRC), University of Galway, Galway, Ireland
| | - Phillip Stafford
- Arizona State University, School of Life Sciences, Biodesign Institute, Arizona, USA
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster (AGRC), University of Galway, Galway, Ireland.
- Aquila Bioscience, University of Galway, Galway, Ireland.
| |
Collapse
|
4
|
Di Filippo ES, Chiappalupi S, Falone S, Dolo V, Amicarelli F, Marchianò S, Carino A, Mascetti G, Valentini G, Piccirillo S, Balsamo M, Vukich M, Fiorucci S, Sorci G, Fulle S. The MyoGravity project to study real microgravity effects on human muscle precursor cells and tissue. NPJ Microgravity 2024; 10:92. [PMID: 39362881 PMCID: PMC11450100 DOI: 10.1038/s41526-024-00432-1] [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: 08/15/2023] [Accepted: 09/22/2024] [Indexed: 10/05/2024] Open
Abstract
Microgravity (µG) experienced during space flights promotes adaptation in several astronauts' organs and tissues, with skeletal muscles being the most affected. In response to reduced gravitational loading, muscles (especially, lower limb and antigravity muscles) undergo progressive mass loss and alteration in metabolism, myofiber size, and composition. Skeletal muscle precursor cells (MPCs), also known as satellite cells, are responsible for the growth and maintenance of muscle mass in adult life as well as for muscle regeneration following damage and may have a major role in µG-induced muscle wasting. Despite the great relevance for astronaut health, very few data are available about the effects of real µG on human muscles. Based on the MyoGravity project, this study aimed to analyze: (i) the cellular and transcriptional alterations induced by real µG in human MPCs (huMPCs) and (ii) the response of human skeletal muscle to normal gravitational loading after prolonged exposure to µG. We evaluated the transcriptomic changes induced by µG on board the International Space Station (ISS) in differentiating huMPCs isolated from Vastus lateralis muscle biopsies of a pre-flight astronaut and an age- and sex-matched volunteer, in comparison with the same cells cultured on the ground in standard gravity (1×g) conditions. We found that huMPCs differentiated under real µG conditions showed: (i) upregulation of genes related to cell adhesion, plasma membrane components, and ion transport; (ii) strong downregulation of genes related to the muscle contraction machinery and sarcomere organization; and (iii) downregulation of muscle-specific microRNAs (myomiRs). Moreover, we had the unique opportunity to analyze huMPCs and skeletal muscle tissue of the same astronaut before and 30 h after a long-duration space flight on board the ISS. Prolonged exposure to real µG strongly affected the biology and functionality of the astronaut's satellite cells, which showed a dramatic reduction of responsiveness to activating stimuli and proliferation rate, morphological changes, and almost inability to fuse into myotubes. RNA-Seq analysis of post- vs. pre-flight muscle tissue showed that genes involved in muscle structure and remodeling are promptly activated after landing following a long-duration space mission. Conversely, genes involved in the myelination process or synapse and neuromuscular junction organization appeared downregulated. Although we have investigated only one astronaut, these results point to a prompt readaptation of the skeletal muscle mechanical components to the normal gravitational loading, but the inability to rapidly recover the physiological muscle myelination/innervation pattern after landing from a long-duration space flight. Together with the persistent functional deficit observed in the astronaut's satellite cells after prolonged exposure to real µG, these results lead us to hypothesize that a condition of inefficient regeneration is likely to occur in the muscles of post-flight astronauts following damage.
Collapse
Affiliation(s)
- Ester Sara Di Filippo
- Department of Neuroscience Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
- Interuniversity Institute of Myology (IIM), 06132, Perugia, Italy
| | - Sara Chiappalupi
- Interuniversity Institute of Myology (IIM), 06132, Perugia, Italy
- Department Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), 34127, Trieste, Italy
| | - Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvia Marchianò
- Department Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
| | - Adriana Carino
- Department Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
| | | | | | | | - Michele Balsamo
- Kayser Italia S.r.l, Via di Popogna, 501, 57128, Livorno, Italy
| | - Marco Vukich
- European Space Agency, Keplerlaan 1, NL-2200, AG, Noordwijk, The Netherlands
| | - Stefano Fiorucci
- Department Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Interuniversity Institute of Myology (IIM), 06132, Perugia, Italy
- Department Medicine and Surgery, University of Perugia, 06132, Perugia, Italy
- Consorzio Interuniversitario Biotecnologie (CIB), 34127, Trieste, Italy
| | - Stefania Fulle
- Department of Neuroscience Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.
- Interuniversity Institute of Myology (IIM), 06132, Perugia, Italy.
| |
Collapse
|
5
|
Poon CCW, Au-Yeung C, Wong KY, Chan Z, Zhou LP, Li G, Wang Y, Zhang Y, Wong MS. Icariin promotes cell adhesion for osteogenesis in bone marrow stromal cells via binding to integrin α5β1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155887. [PMID: 39067311 DOI: 10.1016/j.phymed.2024.155887] [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: 09/11/2023] [Revised: 06/05/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND PURPOSE Icariin, an 8-prenylated flavonoid glycoside, is an anabolic agent that could exert rapid estrogenic actions via ligand-independent activation of estrogen receptor alpha (ERα) in osteoblastic cells to promote osteogenesis. However, relatively little is known about its direct cellular target, its protective effects, and cell adhesion activities in bone marrow stromal cells (BMSCs) against microgravity. In the present study, the effects of icariin on osteogenesis and cell adhesion under microgravity were examined with the involvement of integrin receptor α5β1, connexin 43, and CAMs. STUDY DESIGN AND METHODS Icariin was orally administered to 6-month-old ovariectomized (OVX) Sprague-Dawley (SD) rats for 3 months through daily intake of phytoestrogen-free rodent diets containing icariin at 2 different dosages (50 and 500 ppm). BMSCs were harvested for experiments and RNA-sequencing analysis to examine the mechanism of action of icariin and its direct cellular target in stimulating osteogenesis. RESULTS The results revealed that icariin induced the expression of cell adhesion molecules (CAMs) and protected against microgravity-induced disruption of actin cytoskeleton and the loss of osteogenic activities in BMSCs through the activation of connexin-43 (Cx43) and Ras homolog family member A (RhoA) and Rac family small GTPase 1 (Rac1)-mediated signaling pathways. Computerized molecular docking techniques and the competitive solid-phase binding ELISA assay confirmed that icariin could be a direct ligand of integrin alpha 5 beta 1 (α5β1), and it could also increase the protein expression of integrin α5β1 for mechanosensing. CONCLUSION Our findings suggest that icariin could directly activate cell adhesion signaling by binding to integrin α5β1, which opens up new avenues for the development of integrin α5β1 ligand as an agent to protect against unloading-induced bone loss.
Collapse
Affiliation(s)
- Christina Chui-Wa Poon
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China.; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Chun Au-Yeung
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Ka-Ying Wong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Zoe Chan
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Li-Ping Zhou
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China
| | - Gang Li
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China
| | - Yan Wang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, PR China
| | - Yan Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Man-Sau Wong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China.; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, PR China.; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PR China..
| |
Collapse
|
6
|
Mair DB, Tsui JH, Higashi T, Koenig P, Dong Z, Chen JF, Meir JU, Smith AST, Lee PHU, Ahn EH, Countryman S, Sniadecki NJ, Kim DH. Spaceflight-induced contractile and mitochondrial dysfunction in an automated heart-on-a-chip platform. Proc Natl Acad Sci U S A 2024; 121:e2404644121. [PMID: 39312653 PMCID: PMC11459163 DOI: 10.1073/pnas.2404644121] [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: 03/05/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
With current plans for manned missions to Mars and beyond, the need to better understand, prevent, and counteract the harmful effects of long-duration spaceflight on the body is becoming increasingly important. In this study, an automated heart-on-a-chip platform was flown to the International Space Station on a 1-mo mission during which contractile cardiac function was monitored in real-time. Upon return to Earth, engineered human heart tissues (EHTs) were further analyzed with ultrastructural imaging and RNA sequencing to investigate the impact of prolonged microgravity on cardiomyocyte function and health. Spaceflight EHTs exhibited significantly reduced twitch forces, increased incidences of arrhythmias, and increased signs of sarcomere disruption and mitochondrial damage. Transcriptomic analyses showed an up-regulation of genes and pathways associated with metabolic disorders, heart failure, oxidative stress, and inflammation, while genes related to contractility and calcium signaling showed significant down-regulation. Finally, in silico modeling revealed a potential link between oxidative stress and mitochondrial dysfunction that corresponded with RNA sequencing results. This represents an in vitro model to faithfully reproduce the adverse effects of spaceflight on three-dimensional (3D)-engineered heart tissue.
Collapse
Affiliation(s)
- Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jonathan H. Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Ty Higashi
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
| | - Paul Koenig
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jeffrey F. Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Jessica U. Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX77058
| | - Alec S. T. Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA98195
| | - Peter H. U. Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI02912
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD21205
| | - Stefanie Countryman
- BioServe Space Technologies, Ann and HJ Smead Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO80303
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA98195
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Bioengineering, University of Washington, Seattle, WA98195
- Center for Cardiovascular Biology, University of Washington, Seattle, WA98109
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA98109
- Department of Medicine, Johns Hopkins University, Baltimore, MD21205
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
7
|
Igbineweka NE, van Loon JJWA. Gene-environmental influence of space and microgravity on red blood cells with sickle cell disease. NPJ Genom Med 2024; 9:44. [PMID: 39349487 PMCID: PMC11442622 DOI: 10.1038/s41525-024-00427-7] [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: 07/20/2023] [Accepted: 09/09/2024] [Indexed: 10/02/2024] Open
Abstract
A fundamental question in human biology and for hematological disease is how do complex gene-environment interactions lead to individual disease outcome? This is no less the case for sickle cell disease (SCD), a monogenic disorder of Mendelian inheritance, both clinical course, severity, and treatment response, is variable amongst affected individuals. New insight and discovery often lie between the intersection of seemingly disparate disciplines. Recently, opportunities for space medicine have flourished and have offered a new paradigm for study. Two recent Nature papers have shown that hemolysis and oxidative stress play key mechanistic roles in erythrocyte pathogenesis during spaceflight. This paper reviews existing genetic and environmental modifiers of the sickle cell disease phenotype. It reviews evidence for erythrocyte pathology in microgravity environments and demonstrates why this may be relevant for the unique gene-environment interaction of the SCD phenotype. It also introduces the hematology and scientific community to methodological tools for evaluation in space and microgravity research. The increasing understanding of space biology may yield insight into gene-environment influences and new treatment paradigms in SCD and other hematological disease phenotypes.
Collapse
Affiliation(s)
- Norris E Igbineweka
- Imperial College London, Centre for Haematology, Department of Immunology & Inflammation, Commonwealth Building, Hammersmith Campus, Du Cane, London, W12 0NN, UK.
- Department of Haematology, King's College Hospital NHS Foundation Trust Denmark Hill, SE5 9RS, London, UK.
| | - Jack J W A van Loon
- Dutch Experiment Support Center (DESC), Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam Bone Center (ABC), Amsterdam UMC Location VU University Medical Center (VUmc) & Academic Centre for Dentistry Amsterdam (ACTA), Gustav Mahlerlaan 3004, 1081, LA Amsterdam, The Netherlands
- European Space Agency (ESA), European Space Research and Technology Centre (ESTEC), TEC-MMG, Keplerlaan 1, 2201, AZ Noordwijk, The Netherlands
| |
Collapse
|
8
|
Murali A, Sarkar RR. Dynamic cellular responses to gravitational forces: Exploring the impact on white blood cell(s). BIOMICROFLUIDICS 2024; 18:054112. [PMID: 39445310 PMCID: PMC11495877 DOI: 10.1063/5.0216617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024]
Abstract
In recent years, the allure of space exploration and human spaceflight has surged, yet the effects of microgravity on the human body remain a significant concern. Immune and red blood cells rely on hematic or lymphatic streams as their primary means of transportation, posing notable challenges under microgravity conditions. This study sheds light on the intricate dynamics of cell behavior when suspended in bio-fluid under varying gravitational forces. Utilizing the dissipative particle dynamics approach, blood and white blood cells were modeled, with gravity applied as an external force along the vertical axis, ranging from 0 to 2 g in parameter sweeps. The results revealed discernible alterations in the cell shape and spatial alignment in response to gravity, quantified through metrics such as elongation and deformation indices, pitch angle, and normalized center of mass. Statistical analysis using the Mann-Whitney U test underscored clear distinctions between microgravity (<1 g) and hypergravity (>1 g) samples compared to normal gravity (1 g). Furthermore, the examination of forces exerted on the solid, including drag, shear stress, and solid forces, unveiled a reduction in the magnitude as the gravitational force increased. Additional analysis through dimensionless numbers unveiled the dominance of capillary and gravitational forces, which impacted cell velocity, leading to closer proximity to the wall and heightened viscous interaction with surrounding fluid particles. These interactions prompted shape alterations and reduced white blood cell area while increasing red blood cells. This study represents an effort in comprehending the effects of gravity on blood cells, offering insights into the intricate interplay between cellular dynamics and gravitational forces.
Collapse
Affiliation(s)
| | - Ram Rup Sarkar
- Author to whom correspondence should be addressed:. Telephone: +91-20-2590 3040; +91-20-2590 2161
| |
Collapse
|
9
|
Cotter JA, Plaza-Florido A, Adams GR, Haddad F, Scott JM, Everett M, Ploutz-Snyder L, Radom-Aizik S. Exercise Training Attenuates the Muscle Mitochondria Genomic Response to Bed Rest. Med Sci Sports Exerc 2024; 56:1615-1622. [PMID: 38650118 PMCID: PMC11326991 DOI: 10.1249/mss.0000000000003457] [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] [Indexed: 04/25/2024]
Abstract
PURPOSE Exercise training during the National Aeronautics and Space Administration 70-d bed rest study effectively counteracted the decline in aerobic capacity, muscle mass, strength, and endurance. We aimed to characterize the genomic response of the participants' vastus lateralis on day 64 of bed rest with and without exercise countermeasures. METHODS Twenty-two healthy young males were randomized into three groups: 1) bed rest only ( n = 7), 2) bed rest + aerobic (6 d·wk -1 ) and resistance training (3 d·wk -1 ) on standard equipment ( n = 7), and 3) bed rest + aerobic and resistance training using a flywheel device ( n = 8). The vastus lateralis gene and microRNA microarrays were analyzed using GeneSpring GX 14.9.1 (Agilent Technologies, Palo Alto, CA). RESULTS Bed rest significantly altered the expression of 2113 annotated genes in at least one out of the three study groups (fold change (FC) > 1.2; P < 0.05). Interaction analysis revealed that exercise attenuated the bed rest effect of 511 annotated genes (FC = 1.2, P < 0.05). In the bed rest only group, a predominant downregulation of genes was observed, whereas in the two exercise groups, there was a notable attenuation or reversal of this effect, with no significant differences between the two exercise modalities. Enrichment analysis identified functional categories and gene pathways, many of them related to the mitochondria. In addition, bed rest significantly altered the expression of 35 microRNAs (FC > 1.2, P < 0.05) with no difference between the three groups. Twelve are known to regulate some of the mitochondrial-related genes that were altered following bed rest. CONCLUSIONS Mitochondrial gene expression was a significant component of the molecular response to long-term bed rest. Although exercise attenuated the FC in the downregulation of many genes, it did not completely counteract all the molecular consequences.
Collapse
Affiliation(s)
- Joshua A. Cotter
- Pediatric Exercise and Genomics Research Center, School of Medicine, University of California, Irvine, CA
- Physiology of EXercise and Sport (PEXS) Laboratory, Department of Kinesiology, California State University, Long Beach, CA
| | - Abel Plaza-Florido
- Pediatric Exercise and Genomics Research Center, School of Medicine, University of California, Irvine, CA
| | - Gregory R. Adams
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA
| | - Fadia Haddad
- Pediatric Exercise and Genomics Research Center, School of Medicine, University of California, Irvine, CA
| | - Jessica M. Scott
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Meghan Everett
- National Aeronautics and Space Administration (NASA), Houston, TX
| | | | - Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, School of Medicine, University of California, Irvine, CA
| |
Collapse
|
10
|
Kim S, Ayan B, Shayan M, Rando TA, Huang NF. Skeletal muscle-on-a-chip in microgravity as a platform for regeneration modeling and drug screening. Stem Cell Reports 2024; 19:1061-1073. [PMID: 39059375 PMCID: PMC11368695 DOI: 10.1016/j.stemcr.2024.06.010] [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: 12/29/2023] [Revised: 06/22/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Microgravity has been shown to lead to both muscle atrophy and impaired muscle regeneration. The purpose was to study the efficacy of microgravity to model impaired muscle regeneration in an engineered muscle platform and then to demonstrate the feasibility of performing drug screening in this model. Engineered human muscle was launched to the International Space Station National Laboratory, where the effect of microgravity exposure for 7 days was examined by transcriptomics and proteomics approaches. Gene set enrichment analysis of engineered muscle cultured in microgravity, compared to normal gravity conditions, highlighted a metabolic shift toward lipid and fatty acid metabolism, along with increased apoptotic gene expression. The addition of pro-regenerative drugs, insulin-like growth factor-1 (IGF-1) and a 15-hydroxyprostaglandin dehydrogenase inhibitor (15-PGDH-i), partially inhibited the effects of microgravity. In summary, microgravity mimics aspects of impaired myogenesis, and the addition of these drugs could partially inhibit the effects induced by microgravity.
Collapse
Affiliation(s)
- Soochi Kim
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Bugra Ayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA 94304, USA.
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto, Health Care System, Palo Alto, CA 94304, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
11
|
Jeyaraman M, Ramasubramanian S, Yadav S, Jeyaraman N. Exploring New Horizons: Advancements in Cartilage Tissue Engineering Under Space Microgravity. Cureus 2024; 16:e66224. [PMID: 39238750 PMCID: PMC11374578 DOI: 10.7759/cureus.66224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Novel investigations of how microgravity affects cellular and tissue development have recently been made possible by the multidisciplinary fusion of tissue engineering and space science. This review examines the intersection of cartilage tissue engineering (CTE) and space science, focusing on how microgravity affects cartilage development. Space microgravity induces distinct physiological changes in chondrocytes, including a 20-30% increase in cell diameter, a 1.5- to 2-fold increase in proliferation rates, and up to 3-fold increases in chondrogenic markers such as SOX9 and collagen type II. These cellular alterations impact extracellular matrix composition and tissue structure. Space-optimized bioreactors using dynamic culture methods replicate physiological conditions and enhance tissue growth, but the absence of gravity raises concerns about the mechanical properties of engineered cartilage. Key research areas include the role of growth factors in cartilage development under microgravity, biocompatibility and degradation of scaffold materials in space, and in situ experiments on space stations. This review highlights the opportunities and challenges in leveraging microgravity for CTE advancements, emphasizing the need for continued research to harness space environments for therapeutic applications in cartilage regeneration. The multidisciplinary fusion of tissue engineering and space science opens novel avenues for understanding and improving cartilage tissue engineering, with significant implications for the future of biomedical applications in space and on Earth.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Clinical Research, Virginia Tech India, Dr MGR Educational and Research Institute, Chennai, IND
- Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, IND
| | | | - Sankalp Yadav
- Medicine, Shri Madan Lal Khurana Chest Clinic, New Delhi, IND
| | - Naveen Jeyaraman
- Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, IND
| |
Collapse
|
12
|
Pacelli C, Ferranti F, Del Bianco M. Special Issue: 'Advances in Space Biology'. Life (Basel) 2024; 14:931. [PMID: 39202673 PMCID: PMC11355448 DOI: 10.3390/life14080931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/12/2024] [Indexed: 09/03/2024] Open
Abstract
As we enter a new era of space exploration, space biology is at the forefront of both robotic and human space programs [...].
Collapse
Affiliation(s)
- Claudia Pacelli
- Italian Space Agency, Via del Politecnico s.n.c., 00133 Rome, Italy
- Centre for Space Life Sciences, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Francesca Ferranti
- Italian Space Agency, Via del Politecnico s.n.c., 00133 Rome, Italy
- Centre for Space Life Sciences, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Marta Del Bianco
- Italian Space Agency, Via del Politecnico s.n.c., 00133 Rome, Italy
- Centre for Space Life Sciences, Viale Regina Elena, 299, 00161 Rome, Italy
| |
Collapse
|
13
|
Bakr MM, Caswell GM, Hussein H, Shamel M, Al-Ankily MM. Considerations for oral and dental tissues in holistic care during long-haul space flights. Front Physiol 2024; 15:1406631. [PMID: 39055690 PMCID: PMC11269229 DOI: 10.3389/fphys.2024.1406631] [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: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024] Open
Abstract
The health of astronauts during and after the return from long-haul space missions is paramount. There is plethora of research in the literature about the medical side of astronauts' health, however, the dental and oral health of the space crew seem to be overlooked with limited information in the literature about the effects of the space environment and microgravity on the oral and dental tissues. In this article, we shed some light on the latest available research related to space dentistry and provide some hypotheses that could guide the directions of future research and help maintain the oral health of space crews. We also promote for the importance of regenerative medicine and dentistry as well highlight the opportunities available in the expanding field of bioprinting/biomanufacturing through utilizing the effects of microgravity on stem cells culture techniques. Finally, we provide recommendations for adopting a multidisciplinary approach for oral healthcare during long-haul space flights.
Collapse
Affiliation(s)
- Mahmoud M. Bakr
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
| | | | - Habiba Hussein
- Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Mohamed Shamel
- Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | | |
Collapse
|
14
|
Rabiee S, Zaboli S, Sammak AS, Mohebbi A. Effects of Simulated Microgravity on Rat Reproductive System: Potential Benefits of Vitamin D3 Intervention. Reprod Sci 2024; 31:1973-1982. [PMID: 38600415 DOI: 10.1007/s43032-024-01508-9] [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: 08/31/2023] [Accepted: 03/01/2024] [Indexed: 04/12/2024]
Abstract
Gravity in space can have a negative impact on the reproductive system. Given that the reproductive system is one of vitamin D's objectives, this study will use a simulated microgravity model to evaluate its impact on the rat reproductive system.Twenty-two male Wistar rats were allocated into four groups at random. Under microgravity circumstances, the rats were housed in both special and standard cages. Each group was then separated into two subgroups, one of which received vitamin D3 and the other did not. Blood was drawn twice to determine blood levels of vitamin D3, LH, FSH, and testosterone. Rat testes were isolated for histological analysis, as well as a piece of epididymis for sperm count and morphological examination.Microgravity had a detrimental effect on testicular tissue, resulting in lower serum levels of LH and testosterone (p-value < 0.001). Spermatogenesis was largely inhibited under microgravity. During microgravity conditions, however, vitamin D3 had a good effect on testicular structure, and the total number of sperm. Simulated microgravity affects the male reproductive system, compromising testicular morphology, sperm parameters, and hormonal balance. However, this study shows that vitamin D3 supplementation can act as a preventative strategy, minimizing the negative consequences of microgravity. The beneficial effect of vitamin D3 on testicular health and sperm quality implies that it may be useful in protecting male reproductive function in space-related situations.
Collapse
Affiliation(s)
- Saeed Rabiee
- Department of Biology, Parand Branch, Islamic Azad University, Parand Branch, Tehran, 3761396361, Iran
| | - Shiva Zaboli
- Department of Biology, Parand Branch, Islamic Azad University, Parand Branch, Tehran, 3761396361, Iran
| | | | - Alireza Mohebbi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, 4934174515, Iran.
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Ouchi T, Kono K, Satou R, Kurashima R, Yamaguchi K, Kimura M, Shibukawa Y. Upregulation of Amy1 in the salivary glands of mice exposed to a lunar gravity environment using the multiple artificial gravity research system. Front Physiol 2024; 15:1417719. [PMID: 38989048 PMCID: PMC11233762 DOI: 10.3389/fphys.2024.1417719] [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: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction: Space is a unique environment characterized by isolation from community life and exposure to circadian misalignment, microgravity, and space radiation. These multiple differences from those experienced on the earth may cause systemic and local tissue stress. Autonomic nerves, including sympathetic and parasympathetic nerves, regulate functions in multiple organs. Saliva is secreted from the salivary gland, which is regulated by autonomic nerves, and plays several important roles in the oral cavity and digestive processes. The balance of the autonomic nervous system in the seromucous glands, such as the submandibular glands, precisely controls serous and mucous saliva. Psychological stress, radiation damage, and other triggers can cause an imbalance in salivary secretion systems. A previous study reported that amylase is a stress marker in behavioral medicine and space flight crews; however, the detailed mechanisms underlying amylase regulation in the space environment are still unknown. Methods: In this study, we aimed to elucidate how lunar gravity (1/6 g) changes mRNA expression patterns in the salivary gland. Using a multiple artificial gravity research system during space flight in the International Space Station, we studied the effects of two different gravitational levels, lunar and Earth gravity, on the submandibular glands of mice. All mice survived, returned to Earth from space, and their submandibular glands were collected 2 days after landing. Results: We found that lunar gravity induced the expression of the salivary amylase gene Amy1; however, no increase in Aqp5 and Ano1, which regulate water secretion, was observed. In addition, genes involved in the exocrine system, such as vesicle-associated membrane protein 8 (Vamp8) and small G proteins, including Rap1 and Rab families, were upregulated under lunar gravity. Conclusion: These results imply that lunar gravity upregulates salivary amylase secretion via Rap/Rab signaling and exocytosis via Vamp8. Our study highlights Amy1 as a potential candidate marker for stress regulation in salivary glands in the lunar gravity environment.
Collapse
Affiliation(s)
- Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Kyosuke Kono
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Ryouichi Satou
- Department of Epidemiology and Public Health, Tokyo Dental College, Tokyo, Japan
| | - Ryuya Kurashima
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Koji Yamaguchi
- NeSTRA (Next-Generation Space System Technology Research Association), Yokohama, Japan
| | - Maki Kimura
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | | |
Collapse
|
16
|
Lee Satcher R, Fiedler B, Ghali A, Dirschl DR. Effect of Spaceflight and Microgravity on the Musculoskeletal System: A Review. J Am Acad Orthop Surg 2024; 32:535-541. [PMID: 38652883 DOI: 10.5435/jaaos-d-23-00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
With National Aeronautics and Space Administration's plans for longer distance, longer duration spaceflights such as missions to Mars and the surge in popularity of space tourism, the need to better understand the effects of spaceflight on the musculoskeletal system has never been more present. However, there is a paucity of information on how spaceflight affects orthopaedic health. This review surveys existing literature and discusses the effect of spaceflight on each aspect of the musculoskeletal system. Spaceflight reduces bone mineral density at rapid rates because of multiple mechanisms. While this seems to be recoverable upon re-exposure to gravity, concern for fracture in spaceflight remains as microgravity impairs bone strength and fracture healing. Muscles, tendons, and entheses similarly undergo microgravity adaptation. These changes result in decreased muscle mass, increased tendon laxity, and decreased enthesis stiffness, thus decreasing the strength of the muscle-tendon-enthesis unit with variable recovery upon gravity re-exposure. Spaceflight also affects joint health; unloading of the joints facilitates changes that thin and atrophy cartilage similar to arthritic phenotypes. These changes are likely recoverable upon return to gravity with exercise. Multiple questions remain regarding effects of longer duration flights on health and implications of these findings on terrestrial medicine, which should be the target of future research.
Collapse
Affiliation(s)
- Robert Lee Satcher
- From the Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX (Lee Satcher), and the Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX (Fiedler, Ghali, and Dirschl)
| | | | | | | |
Collapse
|
17
|
Masarapu Y, Cekanaviciute E, Andrusivova Z, Westholm JO, Björklund Å, Fallegger R, Badia-I-Mompel P, Boyko V, Vasisht S, Saravia-Butler A, Gebre S, Lázár E, Graziano M, Frapard S, Hinshaw RG, Bergmann O, Taylor DM, Wallace DC, Sylvén C, Meletis K, Saez-Rodriguez J, Galazka JM, Costes SV, Giacomello S. Spatially resolved multiomics on the neuronal effects induced by spaceflight in mice. Nat Commun 2024; 15:4778. [PMID: 38862479 PMCID: PMC11166911 DOI: 10.1038/s41467-024-48916-8] [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: 05/23/2023] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
Impairment of the central nervous system (CNS) poses a significant health risk for astronauts during long-duration space missions. In this study, we employed an innovative approach by integrating single-cell multiomics (transcriptomics and chromatin accessibility) with spatial transcriptomics to elucidate the impact of spaceflight on the mouse brain in female mice. Our comparative analysis between ground control and spaceflight-exposed animals revealed significant alterations in essential brain processes including neurogenesis, synaptogenesis and synaptic transmission, particularly affecting the cortex, hippocampus, striatum and neuroendocrine structures. Additionally, we observed astrocyte activation and signs of immune dysfunction. At the pathway level, some spaceflight-induced changes in the brain exhibit similarities with neurodegenerative disorders, marked by oxidative stress and protein misfolding. Our integrated spatial multiomics approach serves as a stepping stone towards understanding spaceflight-induced CNS impairments at the level of individual brain regions and cell types, and provides a basis for comparison in future spaceflight studies. For broader scientific impact, all datasets from this study are available through an interactive data portal, as well as the National Aeronautics and Space Administration (NASA) Open Science Data Repository (OSDR).
Collapse
Affiliation(s)
- Yuvarani Masarapu
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Zaneta Andrusivova
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jakub O Westholm
- National Bioinformatics Infrastructure Sweden, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, Stockholm, Sweden
| | - Åsa Björklund
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Robin Fallegger
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Pau Badia-I-Mompel
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
- GSK, Cellzome, Heidelberg, Germany
| | - Valery Boyko
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
- Bionetics, Yorktown, VA, USA
| | - Shubha Vasisht
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Amanda Saravia-Butler
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Samrawit Gebre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Enikő Lázár
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Marta Graziano
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Solna, Sweden
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Robert G Hinshaw
- NASA Postdoctoral Program - Oak Ridge Associated Universities, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Olaf Bergmann
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Pharmacology and Toxicology, Department of Pharmacology and Toxicology University Medical Center Goettingen, Goettingen, Germany
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia and Department of Pediatrics, Division of Human Genetics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christer Sylvén
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA, 94035, USA.
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
18
|
Masiero G, Ferrarese G, Perazzolo E, Baraldo M, Nogara L, Tezze C. Custom-made 3D-printed boot as a model of disuse-induced atrophy in murine skeletal muscle. PLoS One 2024; 19:e0304380. [PMID: 38820523 PMCID: PMC11142711 DOI: 10.1371/journal.pone.0304380] [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: 10/14/2023] [Accepted: 05/10/2024] [Indexed: 06/02/2024] Open
Abstract
Skeletal muscle atrophy is characterized by a decrease in muscle mass and strength caused by an imbalance in protein synthesis and degradation. This process naturally occurs upon reduced or absent physical activity, often related to illness, forced bed rest, or unhealthy lifestyles. Currently, no treatment is available for atrophy, and it can only be prevented by overloading exercise, causing severe problems for patients who cannot exercise due to chronic diseases, disabilities, or being bedridden. The two murine models commonly used to induce muscle atrophy are hindlimb suspension and ankle joint immobilization, both of which come with criticalities. The lack of treatments and the relevance of this atrophic process require a unilateral, safe, and robust model to induce muscle atrophy. In this work, we designed and developed a 3D-printed cast to be used for the study of disuse skeletal muscle atrophy. Applying two halves of the cast is non-invasive, producing little to no swelling or skin damage. The application of the cast induces, in 2-weeks immobilized leg, the activation of atrophy-related genes, causing a muscle weight loss up to 25% in the gastrocnemius muscle, and 31% in the soleus muscle of the immobilized leg compared to the control leg. The cross-sectional area of the fibers is decreased by 31% and 34% respectively, with a peculiar effect on fiber types. In the immobilized gastrocnemius, absolute muscle force is reduced by 38%, while normalized force is reduced by 16%. The contralateral leg did not show signs of overload or hypertrophy when compared to free roaming littermates, offering a good internal control over the immobilized limb. Upon removing the cast, the mice effectively recovered mass and force in 3 weeks.
Collapse
Affiliation(s)
- Giulio Masiero
- Department of Biomedical Science, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Giulia Ferrarese
- Department of Biomedical Science, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Eleonora Perazzolo
- Department of Biomedical Science, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Leonardo Nogara
- Department of Biomedical Science, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Caterina Tezze
- Department of Biomedical Science, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
19
|
Blottner D, Moriggi M, Trautmann G, Furlan S, Block K, Gutsmann M, Torretta E, Barbacini P, Capitanio D, Rittweger J, Limper U, Volpe P, Gelfi C, Salanova M. Nitrosative Stress in Astronaut Skeletal Muscle in Spaceflight. Antioxidants (Basel) 2024; 13:432. [PMID: 38671880 PMCID: PMC11047620 DOI: 10.3390/antiox13040432] [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: 02/12/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Long-duration mission (LDM) astronauts from the International Space Station (ISS) (>180 ISS days) revealed a close-to-normal sarcolemmal nitric oxide synthase type-1 (NOS1) immunoexpression in myofibers together with biochemical and quantitative qPCR changes in deep calf soleus muscle. Nitro-DIGE analyses identified functional proteins (structural, metabolic, mitochondrial) that were over-nitrosylated post- vs. preflight. In a short-duration mission (SDM) astronaut (9 ISS days), s-nitrosylation of a nodal protein of the glycolytic flux, specific proteins in tricarboxylic acid (TCA) cycle, respiratory chain, and over-nitrosylation of creatine kinase M-types as signs of impaired ATP production and muscle contraction proteins were seen. S-nitrosylation of serotransferrin (TF) or carbonic anhydrase 3 (CA3b and 3c) represented signs of acute response microgravity muscle maladaptation. LDM nitrosoprofiles reflected recovery of mitochondrial activity, contraction proteins, and iron transporter TF as signs of muscle adaptation to microgravity. Nitrosated antioxidant proteins, alcohol dehydrogenase 5/S-nitrosoglutathione reductase (ADH5/GSNOR), and selenoprotein thioredoxin reductase 1 (TXNRD1) levels indicated signs of altered redox homeostasis and reduced protection from nitrosative stress in spaceflight. This work presents a novel spaceflight-generated dataset on s-nitrosylated muscle protein signatures from astronauts that helps both to better understand the structural and molecular networks associated to muscular nitrosative stress and to design countermeasures to dysfunction and impaired performance control in human spaceflight missions.
Collapse
Affiliation(s)
- Dieter Blottner
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
- NeuroMuscular System and Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Gabor Trautmann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Sandra Furlan
- C.N.R. Neuroscience Institute, I-35121 Padova, Italy;
| | - Katharina Block
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Martina Gutsmann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
| | - Joern Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (J.R.); (U.L.)
| | - Ulrich Limper
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (J.R.); (U.L.)
- Anesthesiology and Intensive Care Medicine, Merheim Medical Center, Witten/Herdecke University, 51109 Cologne, Germany
| | - Pompeo Volpe
- Department of Biomedical Sciences, Università di Padova, I-35121 Padova, Italy;
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.M.); (P.B.); (D.C.); (C.G.)
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Michele Salanova
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (M.S.)
- NeuroMuscular System and Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| |
Collapse
|
20
|
Khan OM, Gasperini W, Necessary C, Jacobs Z, Perry S, Rexroat J, Nelson K, Gamble P, Clements T, DeLeon M, Howard S, Zavala A, Farach-Carson M, Blaber E, Wu D, Satici A, Uzer G. Development and Characterization of a low intensity vibrational system for microgravity studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.20.567870. [PMID: 38045225 PMCID: PMC10690179 DOI: 10.1101/2023.11.20.567870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The advent of extended-duration human spaceflight demands a better comprehension of the physiological impacts of microgravity. One primary concern is the adverse impact on the musculoskeletal system, including muscle atrophy and bone density reduction. Ground-based microgravity simulations have provided insights, with vibrational bioreactors emerging as potential mitigators of these negative effects. Despite the potential they have, the adaptation of vibrational bioreactors for space remains unfulfilled, resulting in a significant gap in microgravity research. This paper introduces the first automated low-intensity vibrational (LIV) bioreactor designed specifically for the International Space Station (ISS) environment. Our research covers the bioreactor's design and characterization, the selection of an optimal linear guide for consistent 1-axis acceleration, a thorough analysis of its thermal and diffusion dynamics, and the pioneering use of BioMed Clear resin for enhanced scaffold design. This advancement sets the stage for more authentic space-based biological studies, vital for ensuring the safety of future space explorations.
Collapse
|
21
|
Jogdand A, Landolina M, Chen Y. Organs in orbit: how tissue chip technology benefits from microgravity, a perspective. FRONTIERS IN LAB ON A CHIP TECHNOLOGIES 2024; 3:1356688. [PMID: 38915901 PMCID: PMC11195915 DOI: 10.3389/frlct.2024.1356688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Tissue chips have become one of the most potent research tools in the biomedical field. In contrast to conventional research methods, such as 2D cell culture and animal models, tissue chips more directly represent human physiological systems. This allows researchers to study therapeutic outcomes to a high degree of similarity to actual human subjects. Additionally, as rocket technology has advanced and become more accessible, researchers are using the unique properties offered by microgravity to meet specific challenges of modeling tissues on Earth; these include large organoids with sophisticated structures and models to better study aging and disease. This perspective explores the manufacturing and research applications of microgravity tissue chip technology, specifically investigating the musculoskeletal, cardiovascular, and nervous systems.
Collapse
Affiliation(s)
- Aditi Jogdand
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| | - Maxwell Landolina
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
22
|
Shen M, Huang X, Zhao Y, Wang Y, Li H, Jiang Z. Human-like acceleration and deceleration control of a robot astronaut floating in a space station. ISA TRANSACTIONS 2024:S0019-0578(24)00097-1. [PMID: 38458904 DOI: 10.1016/j.isatra.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/10/2024]
Abstract
The acceleration and deceleration (AD) motions are the basic motion modes of robot astronauts moving in a space station. Controlling the locomotion of the robot astronaut is very challenging due to the strong nonlinearity of its complex multi-body dynamics in a gravity-free environment. However, after training, humans can move well in space stations by pushing the bulkhead, and the motion mechanism of humans is a good reference for robot astronauts. The contribution of this study is modeling the human AD motion in a microgravity environment and proposing a human-like control method for robot astronauts moving in space stations. Specifically, the movement and contact force data of the human body during AD motion were collected on an air-floating platform. Through human AD modeling analysis, the mechanism of human motion is discovered, and semi-sinusoidal primitives of contact forces are proposed for AD motion. Then, a dynamic guidance model of human AD motion is built to complete motion planning under contact constraints, which is used as the expected model for the AD control of robot astronauts. Benefiting from the force primitives, accurate and safe planning of human-like AD motion can be completed. The characteristics and mechanism of human AD motion have been analyzed from the perspective of optimization. Lastly, based on the proposed dynamic guidance model, the AD motion policy is mapped to the robot astronaut system via a system control method based on the equivalent mapping of dynamic responses (force, velocity and pose). Through comparative analysis with real human motion data and simulation results under different conditions, the proposed AD control method can achieve human-like motion efficiently and stably. Even when confronted with errors in the robot's contact velocities and inertia parameters, the method can significantly reduce the motion errors while ensuring stability.
Collapse
Affiliation(s)
- Minghui Shen
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China
| | - Xiao Huang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China
| | - Yan Zhao
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China
| | - Yunlai Wang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China
| | - Hui Li
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China.
| | - Zhihong Jiang
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China; Key Laboratory of Biomimetic Robots and Systems of Chinese Ministry of Education, Beijing, 100081, China.
| |
Collapse
|
23
|
Schoenrock B, Muckelt PE, Hastermann M, Albracht K, MacGregor R, Martin D, Gunga HC, Salanova M, Stokes MJ, Warner MB, Blottner D. Muscle stiffness indicating mission crew health in space. Sci Rep 2024; 14:4196. [PMID: 38378866 PMCID: PMC10879143 DOI: 10.1038/s41598-024-54759-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Muscle function is compromised by gravitational unloading in space affecting overall musculoskeletal health. Astronauts perform daily exercise programmes to mitigate these effects but knowing which muscles to target would optimise effectiveness. Accurate inflight assessment to inform exercise programmes is critical due to lack of technologies suitable for spaceflight. Changes in mechanical properties indicate muscle health status and can be measured rapidly and non-invasively using novel technology. A hand-held MyotonPRO device enabled monitoring of muscle health for the first time in spaceflight (> 180 days). Greater/maintained stiffness indicated countermeasures were effective. Tissue stiffness was preserved in the majority of muscles (neck, shoulder, back, thigh) but Tibialis Anterior (foot lever muscle) stiffness decreased inflight vs. preflight (p < 0.0001; mean difference 149 N/m) in all 12 crewmembers. The calf muscles showed opposing effects, Gastrocnemius increasing in stiffness Soleus decreasing. Selective stiffness decrements indicate lack of preservation despite daily inflight countermeasures. This calls for more targeted exercises for lower leg muscles with vital roles as ankle joint stabilizers and in gait. Muscle stiffness is a digital biomarker for risk monitoring during future planetary explorations (Moon, Mars), for healthcare management in challenging environments or clinical disorders in people on Earth, to enable effective tailored exercise programmes.
Collapse
Affiliation(s)
- Britt Schoenrock
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany
| | - Paul E Muckelt
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Maria Hastermann
- Experimental and Clinical Research Center (ECRC) and NeuroCure Clinical Research Center (NCRC), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | - Hans-Christian Gunga
- Institute of Physiology, Berlin Center of Space Medicine and Extreme Environments, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, Berlin, Germany
| | - Michele Salanova
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany
| | - Maria J Stokes
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Martin B Warner
- School of Health Sciences, University of Southampton, Southampton, UK
| | - Dieter Blottner
- NeuroMuscular System & Signaling Group, Berlin Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany, Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany, 10115, Berlin, Germany.
| |
Collapse
|
24
|
Davis T, Tabury K, Zhu S, Angeloni D, Baatout S, Benchoua A, Bereiter-Hahn J, Bottai D, Buchheim JI, Calvaruso M, Carnero-Diaz E, Castiglioni S, Cavalieri D, Ceccarelli G, Choukér A, Cialdai F, Ciofani G, Coppola G, Cusella G, Degl'Innocenti A, Desaphy JF, Frippiat JP, Gelinsky M, Genchi G, Grano M, Grimm D, Guignandon A, Hahn C, Hatton J, Herranz R, Hellweg CE, Iorio CS, Karapantsios T, van Loon JJWA, Lulli M, Maier J, Malda J, Mamaca E, Morbidelli L, van Ombergen A, Osterman A, Ovsianikov A, Pampaloni F, Pavezlorie E, Pereda-Campos V, Przybyla C, Puhl C, Rettberg P, Rizzo AM, Robson-Brown K, Rossi L, Russo G, Salvetti A, Santucci D, Sperl M, Tavella S, Thielemann C, Willaert R, Szewczyk N, Monici M. How are cell and tissue structure and function influenced by gravity and what are the gravity perception mechanisms? NPJ Microgravity 2024; 10:16. [PMID: 38341423 DOI: 10.1038/s41526-024-00357-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Progress in mechanobiology allowed us to better understand the important role of mechanical forces in the regulation of biological processes. Space research in the field of life sciences clearly showed that gravity plays a crucial role in biological processes. The space environment offers the unique opportunity to carry out experiments without gravity, helping us not only to understand the effects of gravitational alterations on biological systems but also the mechanisms underlying mechanoperception and cell/tissue response to mechanical and gravitational stresses. Despite the progress made so far, for future space exploration programs it is necessary to increase our knowledge on the mechanotransduction processes as well as on the molecular mechanisms underlying microgravity-induced cell and tissue alterations. This white paper reports the suggestions and recommendations of the SciSpacE Science Community for the elaboration of the section of the European Space Agency roadmap "Biology in Space and Analogue Environments" focusing on "How are cells and tissues influenced by gravity and what are the gravity perception mechanisms?" The knowledge gaps that prevent the Science Community from fully answering this question and the activities proposed to fill them are discussed.
Collapse
Affiliation(s)
- Trent Davis
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Kevin Tabury
- Laboratory of Radiobiology, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shouan Zhu
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Debora Angeloni
- Institute of Biorobotics, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Sarah Baatout
- Laboratory of Radiobiology, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | | | - Juergen Bereiter-Hahn
- Institute for Cell Biology and Neurobiology, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Daniele Bottai
- Department Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Judith-Irina Buchheim
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Marco Calvaruso
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | - Eugénie Carnero-Diaz
- Institute of Systematics, Evolution, Biodiversity, Sorbonne University, NMNH, CNRS, EPHE, UA, Paris, France
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | | | - Gabriele Ceccarelli
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Alexander Choukér
- Laboratory of "Translational Research, Stress & Immunity", Department of Anesthesiology, LMU University Hospital Munich, Munich, Germany
| | - Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Research Division, DSBSC-University of Florence, Florence, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, 56025, Italy
| | - Giuseppe Coppola
- Institute of Applied Science and Intelligent Systems - CNR, Naples, Italy
| | - Gabriella Cusella
- Department of Public Health, Experimental Medicine and Forensic, University of Pavia, Pavia, Italy
| | - Andrea Degl'Innocenti
- Department of Medical Biotechnologies, University of Siena, Italy and Smart Bio-Interfaces, IIT, Pontedera, PI, Italy
| | - Jean-Francois Desaphy
- Department of Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Jean-Pol Frippiat
- Stress, Immunity, Pathogens Laboratory, SIMPA, Université de Lorraine, Nancy, France
| | - Michael Gelinsky
- Centre for Translational Bone, Joint & Soft Tissue Research, TU Dresden, Dresden, Germany
| | - Giada Genchi
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, 56025, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto-von-Guericke-University Magdeburg, Germany & Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Alain Guignandon
- SAINBIOSE, INSERM U1059, Université Jean Monnet, Saint-Etienne, F-42000, France
| | | | - Jason Hatton
- European Space Agency, ESTEC, Noordwijk, The Netherlands
| | - Raúl Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Christine E Hellweg
- Radiation Biology Department, Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | | | | | | | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Jeanette Maier
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht & Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Emina Mamaca
- European and International Affairs Department, Ifremer centre Bretagne, Plouzané, France
| | | | | | - Andreas Osterman
- Max von Pettenkofer Institute, Virology, LMU Munich & DZIF, Partner Site Munich, Munich, Germany
| | - Aleksandr Ovsianikov
- 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria
| | - Francesco Pampaloni
- Buchmann Inst. for Molecular Life Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Elizabeth Pavezlorie
- Ludwig Boltzmann Institute for Traumatology, Research Center in Cooperation with AUVA, Vienna, Austria
| | - Veronica Pereda-Campos
- GSBMS/URU EVOLSAN - Medecine Evolutive, Université Paul Sabatier Toulouse III, Toulouse, France
| | - Cyrille Przybyla
- MARBEC, Université de Montpellier, CNRS, Ifremer, IRD, Palavas les Flots, France
| | - Christopher Puhl
- Space Applications NV/SA for European Space Agency, Noordwijk, The Netherlands
| | - Petra Rettberg
- DLR, Institute of Aerospace Medicine, Research Group Astrobiology, Köln, Germany
| | - Angela Maria Rizzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Kate Robson-Brown
- Department of Engineering Mathematics, and Department of Anthropology and Archaeology, University of Bristol, Bristol, UK
| | - Leonardo Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | - Alessandra Salvetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniela Santucci
- Center for Behavioural Sciences and Mental Health, Istituto Superiore Sanità, Rome, Italy
| | | | - Sara Tavella
- IRCCS Ospedale Policlinico San Martino and University of Genoa, DIMES, Genoa, Italy
| | | | - Ronnie Willaert
- Research Group NAMI and NANO, Vrije Universiteit Brussels, Brussels, Belgium
| | - Nathaniel Szewczyk
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Research Division, DSBSC-University of Florence, Florence, Italy.
| |
Collapse
|
25
|
Melica ME, Cialdai F, La Regina G, Risaliti C, Dafichi T, Peired AJ, Romagnani P, Monici M, Lasagni L. Modeled microgravity unravels the roles of mechanical forces in renal progenitor cell physiology. Stem Cell Res Ther 2024; 15:20. [PMID: 38233961 PMCID: PMC10795253 DOI: 10.1186/s13287-024-03633-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The glomerulus is a highly complex system, composed of different interdependent cell types that are subjected to various mechanical stimuli. These stimuli regulate multiple cellular functions, and changes in these functions may contribute to tissue damage and disease progression. To date, our understanding of the mechanobiology of glomerular cells is limited, with most research focused on the adaptive response of podocytes. However, it is crucial to recognize the interdependence between podocytes and parietal epithelial cells, in particular with the progenitor subset, as it plays a critical role in various manifestations of glomerular diseases. This highlights the necessity to implement the analysis of the effects of mechanical stress on renal progenitor cells. METHODS Microgravity, modeled by Rotary Cell Culture System, has been employed as a system to investigate how renal progenitor cells respond to alterations in the mechanical cues within their microenvironment. Changes in cell phenotype, cytoskeleton organization, cell proliferation, cell adhesion and cell capacity for differentiation into podocytes were analyzed. RESULTS In modeled microgravity conditions, renal progenitor cells showed altered cytoskeleton and focal adhesion organization associated with a reduction in cell proliferation, cell adhesion and spreading capacity. Moreover, mechanical forces appeared to be essential for renal progenitor differentiation into podocytes. Indeed, when renal progenitors were exposed to a differentiative agent in modeled microgravity conditions, it impaired the acquisition of a complex podocyte-like F-actin cytoskeleton and the expression of specific podocyte markers, such as nephrin and nestin. Importantly, the stabilization of the cytoskeleton with a calcineurin inhibitor, cyclosporine A, rescued the differentiation of renal progenitor cells into podocytes in modeled microgravity conditions. CONCLUSIONS Alterations in the organization of the renal progenitor cytoskeleton due to unloading conditions negatively affect the regenerative capacity of these cells. These findings strengthen the concept that changes in mechanical cues can initiate a pathophysiological process in the glomerulus, not only altering podocyte actin cytoskeleton, but also extending the detrimental effect to the renal progenitor population. This underscores the significance of the cytoskeleton as a druggable target for kidney diseases.
Collapse
Affiliation(s)
- Maria Elena Melica
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Gilda La Regina
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Chiara Risaliti
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Tommaso Dafichi
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Anna Julie Peired
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Paola Romagnani
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, 50139, Florence, Italy
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| | - Laura Lasagni
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| |
Collapse
|
26
|
Ibrahim Z, Khan NA, Qaisar R, Saleh MA, Siddiqui R, Al-Hroub HM, Giddey AD, Semreen MH, Soares NC, Elmoselhi AB. Serum multi-omics analysis in hindlimb unloading mice model: Insights into systemic molecular changes and potential diagnostic and therapeutic biomarkers. Heliyon 2024; 10:e23592. [PMID: 38187258 PMCID: PMC10770503 DOI: 10.1016/j.heliyon.2023.e23592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Microgravity, in space travel and prolonged bed rest conditions, induces cardiovascular deconditioning along with skeletal muscle mass loss and weakness. The findings of microgravity research may also aid in the understanding and treatment of human health conditions on Earth such as muscle atrophy, and cardiovascular diseases. Due to the paucity of biomarkers and the unknown underlying mechanisms of cardiovascular and skeletal muscle deconditioning in these environments, there are insufficient diagnostic and preventative measures. In this study, we employed hindlimb unloading (HU) mouse model, which mimics astronauts in space and bedridden patients, to first evaluate cardiovascular and skeletal muscle function, followed by proteomics and metabolomics LC-MS/MS-based analysis using serum samples. Three weeks of unloading caused changes in the function of the cardiovascular system in c57/Bl6 mice, as seen by a decrease in mean arterial pressure and heart weight. Unloading for three weeks also changed skeletal muscle function, causing a loss in grip strength in HU mice and atrophy of skeletal muscle indicated by a reduction in muscle mass. These modifications were partially reversed by a two-week recovery period of reloading condition, emphasizing the significance of the recovery process. Proteomics analysis revealed 12 dysregulated proteins among the groups, such as phospholipid transfer protein, Carbonic anhydrase 3, Parvalbumin alpha, Major urinary protein 20 (Mup20), Thrombospondin-1, and Apolipoprotein C-IV. On the other hand, metabolomics analysis showed altered metabolites among the groups such as inosine, hypoxanthine, xanthosine, sphinganine, l-valine, 3,4-Dihydroxyphenylglycol, and l-Glutamic acid. The joint data analysis revealed that HU conditions mainly impacted pathways such as ABC transporters, complement and coagulation cascades, nitrogen metabolism, and purine metabolism. Overall, our results indicate that microgravity environment induces significant alterations in the function, proteins, and metabolites of these mice. These observations suggest the potential utilization of these proteins and metabolites as novel biomarkers for assessing and mitigating cardiovascular and skeletal muscle deconditioning associated with such conditions.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Naveed A. Khan
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University Edinburgh, EH14 4AS UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Hamza M. Al-Hroub
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Alexander D. Giddey
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohammad Harb Semreen
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av. Padre Cruz, Lisbon, 1649-016, Portugal
- Centre for Toxicogenomics and Human Health (ToxOmics), NOVA School/ Faculdade de Lisboa, Lisbon, Portugal
| | - Adel B. Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
27
|
Smith C, Sim M, Dalla Via J, Levinger I, Duque G. The Interconnection Between Muscle and Bone: A Common Clinical Management Pathway. Calcif Tissue Int 2024; 114:24-37. [PMID: 37922021 DOI: 10.1007/s00223-023-01146-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/26/2023] [Indexed: 11/05/2023]
Abstract
Often observed with aging, the loss of skeletal muscle (sarcopenia) and bone (osteoporosis) mass, strength, and quality, is associated with reduced physical function contributing to falls and fractures. Such events can lead to a loss of independence and poorer quality of life. Physical inactivity (mechanical unloading), especially in older adults, has detrimental effects on the mass and quality of bone as well as muscle, while increases in activity (mechanical loading) have positive effects. Emerging evidence suggests that the relationship between bone and muscle is driven, at least in part, by bone-muscle crosstalk. Bone and muscle are closely linked anatomically, mechanically, and biochemically, and both have the capacity to function with paracrine and endocrine-like action. However, the exact mechanisms involved in this crosstalk remain only partially explored. Given older adults with lower bone mass are more likely to present with impaired muscle function, and vice versa, strategies capable of targeting both bone and muscle are critical. Exercise is the primary evidence-based prevention strategy capable of simultaneously improving muscle and bone health. Unfortunately, holistic treatment plans including exercise in conjunction with other allied health services to prevent or treat musculoskeletal disease remain underutilized. With a focus on sarcopenia and osteoporosis, the aim of this review is to (i) briefly describe the mechanical and biochemical interactions between bone and muscle; (ii) provide a summary of therapeutic strategies, specifically exercise, nutrition and pharmacological approaches; and (iii) highlight a holistic clinical pathway for the assessment and management of sarcopenia and osteoporosis.
Collapse
Affiliation(s)
- Cassandra Smith
- School of Medical and Health Sciences, Nutrition and Health Innovation Research Institute, Edith Cowan University, Joondalup, WA, Australia
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Marc Sim
- School of Medical and Health Sciences, Nutrition and Health Innovation Research Institute, Edith Cowan University, Joondalup, WA, Australia
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Jack Dalla Via
- School of Medical and Health Sciences, Nutrition and Health Innovation Research Institute, Edith Cowan University, Joondalup, WA, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Gustavo Duque
- Bone, Muscle & Geroscience Research Group, Research Institute of the MUHC, Montreal, QC, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Friedman MA, Buettmann EG, Zeineddine Y, Abraham LB, Hoppock GA, Meas SJ, Zhang Y, Farber CR, Donahue HJ. Genetic variation influences the skeletal response to hindlimb unloading in the eight founder strains of the diversity outbred mouse population. J Orthop Res 2024; 42:134-140. [PMID: 37321985 PMCID: PMC10721729 DOI: 10.1002/jor.25646] [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: 03/14/2023] [Revised: 06/11/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
During disuse, mechanical unloading causes extensive bone loss, decreasing bone volume and strength. Variations in bone mass and risk of osteoporosis are influenced by genetics; however, it remains unclear how genetic variation affects the skeletal response to unloading. We previously found that genetic variation affects the musculoskeletal response to 3 weeks of immobilization in the 8 Jackson Laboratory J:DO founder strains: C57Bl/6J, A/J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HlLtJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ. Hindlimb unloading (HLU) is the best model for simulating local and systemic contributors of disuse and therefore may have a greater impact on bones than immobilization. We hypothesized that genetic variation would affect the response to HLU across the eight founder strains. Mice of each founder strain were placed in HLU for 3 weeks, and the femurs and tibias were analyzed. There were significant HLU and mouse strain interactions on body weight, femur trabecular BV/TV, and femur ultimate force. This indicates that unloading only caused significant catabolic effects in some mouse strains. C57BL/6 J mice were most affected by unloading while other strains were more protected. There were significant HLU and mouse strain interactions on gene expression of genes encoding bone metabolism genes in the tibia. This indicates that unloading only caused significant effects on bone metabolism genes in some mouse strains. Different mouse strains respond to HLU differently, and this can be explained by genetic differences. These results suggest the outbred J:DO mice will be a powerful model for examining the effects of genetics on the skeletal response to HLU.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yue Zhang
- Virginia Commonwealth University, Richmond VA
| | | | | |
Collapse
|
29
|
Garbacki N, Willems J, Neutelings T, Lambert C, Deroanne C, Adrian A, Franz M, Maurer M, De Gieter P, Nusgens B, Colige A. Microgravity triggers ferroptosis and accelerates senescence in the MG-63 cell model of osteoblastic cells. NPJ Microgravity 2023; 9:91. [PMID: 38104197 PMCID: PMC10725437 DOI: 10.1038/s41526-023-00339-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023] Open
Abstract
In space, cells sustain strong modifications of their mechanical environment. Mechanosensitive molecules at the cell membrane regulate mechanotransduction pathways that induce adaptive responses through the regulation of gene expression, post-translational modifications, protein interactions or intracellular trafficking, among others. In the current study, human osteoblastic cells were cultured on the ISS in microgravity and at 1 g in a centrifuge, as onboard controls. RNAseq analyses showed that microgravity inhibits cell proliferation and DNA repair, stimulates inflammatory pathways and induces ferroptosis and senescence, two pathways related to ageing. Morphological hallmarks of senescence, such as reduced nuclear size and changes in chromatin architecture, proliferation marker distribution, tubulin acetylation and lysosomal transport were identified by immunofluorescence microscopy, reinforcing the hypothesis of induction of cell senescence in microgravity during space flight. These processes could be attributed, at least in part, to the regulation of YAP1 and its downstream effectors NUPR1 and CKAP2L.
Collapse
Affiliation(s)
- Nancy Garbacki
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Jérôme Willems
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Thibaut Neutelings
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Charles Lambert
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Christophe Deroanne
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Astrid Adrian
- Airbus Defence and Space, GmbH, 88090, Immenstaad, Germany
| | - Markus Franz
- Airbus Defence and Space, GmbH, 88090, Immenstaad, Germany
| | - Matthias Maurer
- European Space Agency (ESA), European Astronaut Centre (EAC), 51147, Cologne, Germany
| | | | - Betty Nusgens
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, 4000, Liège, Belgium.
| |
Collapse
|
30
|
Li K, Desai R, Scott RT, Steele JR, Machado M, Demharter S, Hoarfrost A, Braun JL, Fajardo VA, Sanders LM, Costes SV. Explainable machine learning identifies multi-omics signatures of muscle response to spaceflight in mice. NPJ Microgravity 2023; 9:90. [PMID: 38092777 PMCID: PMC10719374 DOI: 10.1038/s41526-023-00337-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The adverse effects of microgravity exposure on mammalian physiology during spaceflight necessitate a deep understanding of the underlying mechanisms to develop effective countermeasures. One such concern is muscle atrophy, which is partly attributed to the dysregulation of calcium levels due to abnormalities in SERCA pump functioning. To identify potential biomarkers for this condition, multi-omics data and physiological data available on the NASA Open Science Data Repository (osdr.nasa.gov) were used, and machine learning methods were employed. Specifically, we used multi-omics (transcriptomic, proteomic, and DNA methylation) data and calcium reuptake data collected from C57BL/6 J mouse soleus and tibialis anterior tissues during several 30+ day-long missions on the international space station. The QLattice symbolic regression algorithm was introduced to generate highly explainable models that predict either experimental conditions or calcium reuptake levels based on multi-omics features. The list of candidate models established by QLattice was used to identify key features contributing to the predictive capability of these models, with Acyp1 and Rps7 proteins found to be the most predictive biomarkers related to the resilience of the tibialis anterior muscle in space. These findings could serve as targets for future interventions aiming to reduce the extent of muscle atrophy during space travel.
Collapse
Affiliation(s)
- Kevin Li
- KBR, Moffett Field, CA, USA
- NASA Space Life Sciences Training Program, Moffett Field, CA, USA
| | - Riya Desai
- College of Letters and Science, University of California at Davis, Davis, CA, USA
| | - Ryan T Scott
- KBR, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Joel Ricky Steele
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Blue Marble Space, Seattle, WA, USA
| | | | | | | | - Jessica L Braun
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Val A Fajardo
- Department of Kinesiology, Centre for Bone and Muscle Health, Brock University, St. Catharines, Canada
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Blue Marble Space, Seattle, WA, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
| |
Collapse
|
31
|
Zeineddine Y, Friedman MA, Buettmann EG, Abraham LB, Hoppock GA, Donahue HJ. Genetic diversity modulates the physical and transcriptomic response of skeletal muscle to simulated microgravity in male mice. NPJ Microgravity 2023; 9:86. [PMID: 38040743 PMCID: PMC10692100 DOI: 10.1038/s41526-023-00334-8] [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: 05/17/2023] [Accepted: 11/09/2023] [Indexed: 12/03/2023] Open
Abstract
Developments in long-term space exploration necessitate advancements in countermeasures against microgravity-induced skeletal muscle loss. Astronaut data shows considerable variation in muscle loss in response to microgravity. Previous experiments suggest that genetic background influences the skeletal muscle response to unloading, but no in-depth analysis of genetic expression has been performed. Here, we placed eight, male, inbred founder strains of the diversity outbred mice (129S1/SvImJ, A/J, C57BL/6J, CAST/EiJ, NOD/ShiLtJ, NZO/HILtJ, PWK/PhJ, and WSB/EiJ) in simulated microgravity (SM) via hindlimb unloading for three weeks. Body weight, muscle morphology, muscle strength, protein synthesis marker expression, and RNA expression were collected. A/J and CAST/EiJ mice were most susceptible to SM-induced muscle loss, whereas NOD/ShiLtJ mice were the most protected. In response to SM, A/J and CAST/EiJ mice experienced reductions in body weight, muscle mass, muscle volume, and muscle cross-sectional area. A/J mice had the highest number of differentially expressed genes (68) and associated gene ontologies (328). Downregulation of immunological gene ontologies and genes encoding anabolic immune factors suggest that immune dysregulation contributes to the response of A/J mice to SM. Several muscle properties showed significant interactions between SM and mouse strain and a high degree of heritability. These data imply that genetic background plays a role in the degree of muscle loss in SM and that more individualized programs should be developed for astronauts to protect their skeletal muscles against microgravity on long-term missions.
Collapse
Affiliation(s)
- Yasmina Zeineddine
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael A Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Evan G Buettmann
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Lovell B Abraham
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Gabriel A Hoppock
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Henry J Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
32
|
Ha P, Kwak JH, Zhang Y, Shi J, Tran L, Liu TP, Pan HC, Lee S, Kim JK, Chen E, Shirazi-Fard Y, Stodieck LS, Lin A, Zheng Z, Dong SN, Zhang X, Wu BM, Ting K, Soo C. Bisphosphonate conjugation enhances the bone-specificity of NELL-1-based systemic therapy for spaceflight-induced bone loss in mice. NPJ Microgravity 2023; 9:75. [PMID: 37723136 PMCID: PMC10507033 DOI: 10.1038/s41526-023-00319-7] [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: 04/18/2022] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).
Collapse
Affiliation(s)
- Pin Ha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jin Hee Kwak
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Forsyth Institute, Cambridge, MA, 02142, USA
| | - Jiayu Shi
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luan Tran
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy Pan Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin-Chuan Pan
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Samantha Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jong Kil Kim
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eric Chen
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yasaman Shirazi-Fard
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Louis S Stodieck
- BioServe Space Technologies and Aerospace Engineering Sciences, University of Colorado, Boulder, CO, 80303, USA
| | - Andy Lin
- Office of Advanced Research Computing, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Stella Nuo Dong
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin M Wu
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Kang Ting
- Forsyth Institute, Cambridge, MA, 02142, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
33
|
Parafati M, Giza S, Shenoy TS, Mojica-Santiago JA, Hopf M, Malany LK, Platt D, Moore I, Jacobs ZA, Kuehl P, Rexroat J, Barnett G, Schmidt CE, McLamb WT, Clements T, Coen PM, Malany S. Human skeletal muscle tissue chip autonomous payload reveals changes in fiber type and metabolic gene expression due to spaceflight. NPJ Microgravity 2023; 9:77. [PMID: 37714852 PMCID: PMC10504373 DOI: 10.1038/s41526-023-00322-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 08/16/2023] [Indexed: 09/17/2023] Open
Abstract
Microphysiological systems provide the opportunity to model accelerated changes at the human tissue level in the extreme space environment. Spaceflight-induced muscle atrophy experienced by astronauts shares similar physiological changes to muscle wasting in older adults, known as sarcopenia. These shared attributes provide a rationale for investigating molecular changes in muscle cells exposed to spaceflight that may mimic the underlying pathophysiology of sarcopenia. We report the results from three-dimensional myobundles derived from muscle biopsies from young and older adults, integrated into an autonomous CubeLab™, and flown to the International Space Station (ISS) aboard SpaceX CRS-21 as part of the NIH/NASA funded Tissue Chips in Space program. Global transcriptomic RNA-Seq analyses comparing the myobundles in space and on the ground revealed downregulation of shared transcripts related to myoblast proliferation and muscle differentiation. The analyses also revealed downregulated differentially expressed gene pathways related to muscle metabolism unique to myobundles derived from the older cohort exposed to the space environment compared to ground controls. Gene classes related to inflammatory pathways were downregulated in flight samples cultured from the younger cohort compared to ground controls. Our muscle tissue chip platform provides an approach to studying the cell autonomous effects of spaceflight on muscle cell biology that may not be appreciated on the whole organ or organism level and sets the stage for continued data collection from muscle tissue chip experimentation in microgravity. We also report on the challenges and opportunities for conducting autonomous tissue-on-chip CubeLabTM payloads on the ISS.
Collapse
Affiliation(s)
- Maddalena Parafati
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Shelby Giza
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Tushar S Shenoy
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jorge A Mojica-Santiago
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32610, USA
| | - Meghan Hopf
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | | | - Don Platt
- Micro Aerospace Solutions, INC, Melbourne, FL, 32935, USA
| | | | | | - Paul Kuehl
- Space Tango, LLC, Lexington, KY, 40505, USA
| | | | | | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, 32610, USA
| | | | | | - Paul M Coen
- Translational Research Institute, AdventHealth, Orlando, FL, 32804, USA
| | - Siobhan Malany
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
34
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
35
|
Carvajal-Agudelo JD, McNeil A, Franz-Odendaal TA. Effects of simulated microgravity and vibration on osteoblast and osteoclast activity in cultured zebrafish scales. LIFE SCIENCES IN SPACE RESEARCH 2023; 38:39-45. [PMID: 37481306 DOI: 10.1016/j.lssr.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 07/24/2023]
Abstract
Zebrafish cultured scales have been used effectively to study cellular and molecular responses of bone cells. In order to expose zebrafish scales to simulated microgravity (SMG) and/or vibration, we first determined via apoptosis staining whether cells of the scale survive in culture for two days and hence, we restricted our analyses to two-day durations. Next, we measured the effects of SMG and vibration on cell death, osteoclast tartrate-resistant acid phosphatase, and osteoblast alkaline phosphatase activity and on the number of Runx2a positive cells. We found that during the SMG treatment, osteoclast tartrate-resistant acid phosphatase activity increased on average, while the number of Runx2a positive cells decreased significantly. In contrast, SMG exposure caused a decrease in osteoblast activity. The vibration treatment showed an increase, on average, in the osteoblast alkaline phosphatase activity. This study demonstrates the effect of SMG and vibration on zebrafish scales and the effects of SMG on bone cells. We also show that zebrafish scales can be used to examine the effects of SMG on bone maintenance.
Collapse
Affiliation(s)
| | - Alisha McNeil
- Department of Biology, Mount Saint Vincent University, Halifax, NS, B3M 2J6, Canada
| | | |
Collapse
|
36
|
Licata JP, Schwab KH, Har-El YE, Gerstenhaber JA, Lelkes PI. Bioreactor Technologies for Enhanced Organoid Culture. Int J Mol Sci 2023; 24:11427. [PMID: 37511186 PMCID: PMC10380004 DOI: 10.3390/ijms241411427] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
An organoid is a 3D organization of cells that can recapitulate some of the structure and function of native tissue. Recent work has seen organoids gain prominence as a valuable model for studying tissue development, drug discovery, and potential clinical applications. The requirements for the successful culture of organoids in vitro differ significantly from those of traditional monolayer cell cultures. The generation and maturation of high-fidelity organoids entails developing and optimizing environmental conditions to provide the optimal cues for growth and 3D maturation, such as oxygenation, mechanical and fluidic activation, nutrition gradients, etc. To this end, we discuss the four main categories of bioreactors used for organoid culture: stirred bioreactors (SBR), microfluidic bioreactors (MFB), rotating wall vessels (RWV), and electrically stimulating (ES) bioreactors. We aim to lay out the state-of-the-art of both commercial and in-house developed bioreactor systems, their benefits to the culture of organoids derived from various cells and tissues, and the limitations of bioreactor technology, including sterilization, accessibility, and suitability and ease of use for long-term culture. Finally, we discuss future directions for improvements to existing bioreactor technology and how they may be used to enhance organoid culture for specific applications.
Collapse
Affiliation(s)
- Joseph P Licata
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Kyle H Schwab
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yah-El Har-El
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Jonathan A Gerstenhaber
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
37
|
Patel OV, Partridge C, Plaut K. Space Environment Impacts Homeostasis: Exposure to Spaceflight Alters Mammary Gland Transportome Genes. Biomolecules 2023; 13:biom13050872. [PMID: 37238741 DOI: 10.3390/biom13050872] [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: 03/09/2023] [Revised: 04/22/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Membrane transporters and ion channels that play an indispensable role in metabolite trafficking have evolved to operate in Earth's gravity. Dysregulation of the transportome expression profile at normogravity not only affects homeostasis along with drug uptake and distribution but also plays a key role in the pathogenesis of diverse localized to systemic diseases including cancer. The profound physiological and biochemical perturbations experienced by astronauts during space expeditions are well-documented. However, there is a paucity of information on the effect of the space environment on the transportome profile at an organ level. Thus, the goal of this study was to analyze the effect of spaceflight on ion channels and membrane substrate transporter genes in the periparturient rat mammary gland. Comparative gene expression analysis revealed an upregulation (p < 0.01) of amino acid, Ca2+, K+, Na+, Zn2+, Cl-, PO43-, glucose, citrate, pyruvate, succinate, cholesterol, and water transporter genes in rats exposed to spaceflight. Genes associated with the trafficking of proton-coupled amino acids, Mg2+, Fe2+, voltage-gated K+-Na+, cation-coupled chloride, as well as Na+/Ca2+ and ATP-Mg/Pi exchangers were suppressed (p < 0.01) in these spaceflight-exposed rats. These findings suggest that an altered transportome profile contributes to the metabolic modulations observed in the rats exposed to the space environment.
Collapse
Affiliation(s)
- Osman V Patel
- Cell and Molecular Biology Department, Grand Valley State University, Allendale, MI 49401, USA
| | - Charlyn Partridge
- Annis Water Resources Institute, Grand Valley State University, Muskegon, MI 49441, USA
| | - Karen Plaut
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
38
|
Abstract
Bioprinting as an extension of 3D printing offers capabilities for printing tissues and organs for application in biomedical engineering. Conducting bioprinting in space, where the gravity is zero, can enable new frontiers in tissue engineering. Fabrication of soft tissues, which usually collapse under their own weight, can be accelerated in microgravity conditions as the external forces are eliminated. Furthermore, human colonization in space can be supported by providing critical needs of life and ecosystems by 3D bioprinting without relying on cargos from Earth, e.g., by development and long-term employment of living engineered filters (such as sea sponges-known as critical for initiating and maintaining an ecosystem). This review covers bioprinting methods in microgravity along with providing an analysis on the process of shipping bioprinters to space and presenting a perspective on the prospects of zero-gravity bioprinting.
Collapse
Affiliation(s)
- Misagh Rezapour Sarabi
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Savas Tasoglu
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
- Koç University Translational Medicine Research Center (KUTTAM), Koç University, Istanbul, Turkey 34450
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Istanbul, Turkey 34450
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey 34684
| |
Collapse
|
39
|
Dickerson BL, Sowinski R, Kreider RB, Wu G. Impacts of microgravity on amino acid metabolism during spaceflight. Exp Biol Med (Maywood) 2023; 248:380-393. [PMID: 36775855 PMCID: PMC10281620 DOI: 10.1177/15353702221139189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
Spaceflight exerts an extreme and unique influence on human physiology as astronauts are subjected to long-term or short-term exposure to microgravity. During spaceflight, a multitude of physiological changes, including the loss of skeletal muscle mass, bone resorption, oxidative stress, and impaired blood flow, occur, which can affect astronaut health and the likelihood of mission success. In vivo and in vitro metabolite studies suggest that amino acids are among the most affected nutrients and metabolites by microgravity (a weightless condition due to very weak gravitational forces). Moreover, exposure to microgravity alters gut microbial composition, immune function, musculoskeletal health, and consequently amino acid metabolism. Appropriate knowledge of daily protein consumption, with a focus on specific functional amino acids, may offer insight into potential combative and/or therapeutic effects of amino acid consumption in astronauts and space travelers. This will further aid in the successful development of long-term manned space mission and permanent space habitats.
Collapse
Affiliation(s)
- Broderick L Dickerson
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Ryan Sowinski
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Richard B Kreider
- Department of Kinesiology and Sports
Management, Texas A&M University, College Station, TX 77840, USA
| | - Guoyao Wu
- Department of Animal Science and
Faculty of Nutrition, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
40
|
McDonagh F, Cormican M, Morris D, Burke L, Singh NK, Venkateswaran K, Miliotis G. Medical Astro-Microbiology: Current Role and Future Challenges. J Indian Inst Sci 2023; 103:1-26. [PMID: 37362850 PMCID: PMC10082442 DOI: 10.1007/s41745-023-00360-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/03/2023] [Indexed: 06/28/2023]
Abstract
The second and third decades of the twenty-first century are marked by a flourishing of space technology which may soon realise human aspirations of a permanent multiplanetary presence. The prevention, control and management of infection with microbial pathogens is likely to play a key role in how successful human space aspirations will become. This review considers the emerging field of medical astro-microbiology. It examines the current evidence regarding the risk of infection during spaceflight via host susceptibility, alterations to the host's microbiome as well as exposure to other crew members and spacecraft's microbiomes. It also considers the relevance of the hygiene hypothesis in this regard. It then reviews the current evidence related to infection risk associated with microbial adaptability in spaceflight conditions. There is a particular focus on the International Space Station (ISS), as one of the only two crewed objects in low Earth orbit. It discusses the effects of spaceflight related stressors on viruses and the infection risks associated with latent viral reactivation and increased viral shedding during spaceflight. It then examines the effects of the same stressors on bacteria, particularly in relation to changes in virulence and drug resistance. It also considers our current understanding of fungal adaptability in spaceflight. The global public health and environmental risks associated with a possible re-introduction to Earth of invasive species are also briefly discussed. Finally, this review examines the largely unknown microbiology and infection implications of celestial body habitation with an emphasis placed on Mars. Overall, this review summarises much of our current understanding of medical astro-microbiology and identifies significant knowledge gaps. Graphical Abstract
Collapse
Affiliation(s)
- Francesca McDonagh
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Galway, Ireland
| | - Martin Cormican
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Galway, Ireland
- Department of Medical Microbiology, Galway University Hospitals, Galway, Ireland
| | - Dearbháile Morris
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Galway, Ireland
| | - Liam Burke
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Galway, Ireland
| | - Nitin Kumar Singh
- Biotechnology and Planetary Protection Group, NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA USA
| | - Kasthuri Venkateswaran
- Biotechnology and Planetary Protection Group, NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA USA
| | - Georgios Miliotis
- Antimicrobial Resistance and Microbial Ecology Group, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
41
|
Hardy M, Feehan L, Savvides G, Wong J. How controlled motion alters the biophysical properties of musculoskeletal tissue architecture. J Hand Ther 2023; 36:269-279. [PMID: 37029054 DOI: 10.1016/j.jht.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/06/2022] [Indexed: 04/09/2023]
Abstract
INTRODUCTION Movement is fundamental to the normal behaviour of the hand, not only for day-to-day activity, but also for fundamental processes like development, tissue homeostasis and repair. Controlled motion is a concept that hand therapists apply to their patients daily for functional gains, yet the scientific understanding of how this works is poorly understood. PURPOSE OF THE ARTICLE To review the biology of the tissues in the hand that respond to movement and provide a basic science understanding of how it can be manipulated to facilitate better functionThe review outlines the concept of controlled motion and actions across the scales of tissue architecture, highlighting the the role of movement forces in tissue development, homeostasis and repair. The biophysical behaviour of mechanosensitve tissues of the hand such as skin, tendon, bone and cartilage are discussed. CONCLUSION Controlled motion during early healing is a form of controlled stress and can be harnessed to generate appropriate reparative tissues. Understanding the temporal and spatial biology of tissue repair allows therapists to tailor therapies that allow optimal recovery based around progressive biophysical stimuli by movement.
Collapse
Affiliation(s)
- Maureen Hardy
- Past Director Rehab Services and Hand Management Center, St. Dominic Hospital, Jackson, MS, USA
| | - Lynne Feehan
- Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Georgia Savvides
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Manchester Academic Health Science Centre, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jason Wong
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Manchester Academic Health Science Centre, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
42
|
Parafati M, Giza S, Shenoy T, Mojica-Santiago J, Hopf M, Malany L, Platt D, Kuehl P, Moore I, Jacobs Z, Barnett G, Schmidt C, McLamb W, Coen P, Clements T, Malany S. Validation of Human Skeletal Muscle Tissue Chip Autonomous Platform to Model Age-Related Muscle Wasting in Microgravity. RESEARCH SQUARE 2023:rs.3.rs-2631490. [PMID: 37034730 PMCID: PMC10081368 DOI: 10.21203/rs.3.rs-2631490/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Microgravity-induced muscle atrophy experienced by astronauts shares similar physiological changes to muscle wasting experienced by older adults, known as sarcopenia. These shared attributes provide a rationale for investigating microgravity-induced molecular changes in human bioengineered muscle cells that may also mimic the progressive underlying pathophysiology of sarcopenia. Here, we report the results of an experiment that incorporated three-dimensional myobundles derived from muscle biopsies from young and older adults, that were integrated into an autonomous CubeLabâ"¢, and flown to the International Space Station (ISS) aboard SpaceX CRS-21 in December 2020 as part of the NIH/NASA funded Tissue Chips in Space program. Global transcriptomic RNA-Seq analysis comparing the myobundles in space and on the ground revealed downregulation of shared transcripts related to myoblast proliferation and muscle differentiation for those in space. The analysis also revealed differentially expressed gene pathways related to muscle metabolism unique to myobundles derived from the older cohort exposed to the space environment compared to ground controls. Gene classes related to inflammatory pathways were uniquely modulated in flight samples cultured from the younger cohort compared to ground controls. Our muscle tissue chip platform provides a novel approach to studying the cell autonomous effects of microgravity on muscle cell biology that may not be appreciated on the whole organ or organism level and sets the stage for continued data collection from muscle tissue chip experimentation in microgravity. Thus, we also report on the challenges and opportunities for conducting autonomous tissue-on-chip CubeLab TM payloads on the ISS.
Collapse
|
43
|
Iordachescu A, Eisenstein N, Appleby-Thomas G. Space habitats for bioengineering and surgical repair: addressing the requirement for reconstructive and research tissues during deep-space missions. NPJ Microgravity 2023; 9:23. [PMID: 36966158 PMCID: PMC10039948 DOI: 10.1038/s41526-023-00266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/13/2023] [Indexed: 03/27/2023] Open
Abstract
Numerous technical scenarios have been developed to facilitate a human return to the Moon, and as a testbed for a subsequent mission to Mars. Crews appointed with constructing and establishing planetary bases will require a superior level of physical ability to cope with the operational demands. However, the challenging environments of nearby planets (e.g. geological, atmospheric, gravitational conditions) as well as the lengthy journeys through microgravity, will lead to progressive tissue degradation and an increased susceptibility to injury. The isolation, distance and inability to evacuate in an emergency will require autonomous medical support, as well as a range of facilities and specialised equipment to repair tissue damage on-site. Here, we discuss the design requirements of such a facility, in the form of a habitat that would concomitantly allow tissue substitute production, maintenance and surgical implantation, with an emphasis on connective tissues. The requirements for the individual modules and their operation are identified. Several concepts are assessed, including the presence of adjacent wet lab and medical modules supporting the gradual implementation of regenerative biomaterials and acellular tissue substitutes, leading to eventual tissue grafts and, in subsequent decades, potential tissues/organ-like structures. The latter, currently in early phases of development, are assessed particularly for researching the effects of extreme conditions on representative analogues for astronaut health support. Technical solutions are discussed for bioengineering in an isolated planetary environment with hypogravity, from fluid-gel bath suspended manufacture to cryostorage, cell sourcing and on-site resource utilisation for laboratory infrastructure. Surgical considerations are also discussed.
Collapse
Affiliation(s)
- Alexandra Iordachescu
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Consortium for organotypic research on ageing and microgravity, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Cranfield Defence and Security, Cranfield University, Defence Academy of the United Kingdom, Shrivenham, SN6 8LA, United Kingdom.
| | - Neil Eisenstein
- Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Gareth Appleby-Thomas
- Consortium for organotypic research on ageing and microgravity, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
- Cranfield Defence and Security, Cranfield University, Defence Academy of the United Kingdom, Shrivenham, SN6 8LA, United Kingdom
| |
Collapse
|
44
|
Chu WY, Tsia KK. EuniceScope: Low-Cost Imaging Platform for Studying Microgravity Cell Biology. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2023; 4:204-211. [PMID: 38274779 PMCID: PMC10810312 DOI: 10.1109/ojemb.2023.3257991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/13/2023] [Accepted: 03/13/2023] [Indexed: 01/27/2024] Open
Abstract
Microgravity is proven to impact a wide range of human physiology, from stimulating stem cell differentiation to confounding cell health in bones, skeletal muscles, and blood cells. The research in this arena is progressively intensified by the increasing promises of human spaceflights. Considering the limited access to spaceflight, ground-based microgravity-simulating platforms have been indispensable for microgravity-biology research. However, they are generally complex, costly, hard to replicate and reconfigure - hampering the broad adoption of microgravity biology and astrobiology. To address these limitations, we developed a low-cost reconfigurable 3D-printed microscope coined EuniceScope to allow the democratization of astrobiology, especially for educational use. EuniceScope is a compact 2D clinostat system integrated with a modularized brightfield microscope, built upon 3D-printed toolbox. We demonstrated that this compact system offers plausible imaging quality and microgravity-simulating performance. Its high degree of reconfigurability thus holds great promise in the wide dissemination of microgravity-cell-biology research in the broader community, including Science, technology, engineering, and mathematics (STEM) educational and scientific community in the future.
Collapse
Affiliation(s)
- Wing Yan Chu
- University of Hong KongHong Kong
- University of TorontoTorontoONM5SCanada
| | - Kevin K. Tsia
- Department of Electrical and Electronic Engineering, Faculty of EngineeringUniversity of Hong KongHong Kong
| |
Collapse
|
45
|
Blottner D, Moriggi M, Trautmann G, Hastermann M, Capitanio D, Torretta E, Block K, Rittweger J, Limper U, Gelfi C, Salanova M. Space Omics and Tissue Response in Astronaut Skeletal Muscle after Short and Long Duration Missions. Int J Mol Sci 2023; 24:ijms24044095. [PMID: 36835504 PMCID: PMC9962627 DOI: 10.3390/ijms24044095] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The molecular mechanisms of skeletal muscle adaptation to spaceflight are as yet not fully investigated and well understood. The MUSCLE BIOPSY study analyzed pre and postflight deep calf muscle biopsies (m. soleus) obtained from five male International Space Station (ISS) astronauts. Moderate rates of myofiber atrophy were found in long-duration mission (LDM) astronauts (~180 days in space) performing routine inflight exercise as countermeasure (CM) compared to a short-duration mission (SDM) astronaut (11 days in space, little or no inflight CM) for reference control. Conventional H&E scout histology showed enlarged intramuscular connective tissue gaps between myofiber groups in LDM post vs. preflight. Immunoexpression signals of extracellular matrix (ECM) molecules, collagen 4 and 6, COL4 and 6, and perlecan were reduced while matrix-metalloproteinase, MMP2, biomarker remained unchanged in LDM post vs. preflight suggesting connective tissue remodeling. Large scale proteomics (space omics) identified two canonical protein pathways associated to muscle weakness (necroptosis, GP6 signaling/COL6) in SDM and four key pathways (Fatty acid β-oxidation, integrin-linked kinase ILK, Rho A GTPase RHO, dilated cardiomyopathy signaling) explicitly in LDM. The levels of structural ECM organization proteins COL6A1/A3, fibrillin 1, FBN1, and lumican, LUM, increased in postflight SDM vs. LDM. Proteins from tricarboxylic acid, TCA cycle, mitochondrial respiratory chain, and lipid metabolism mostly recovered in LDM vs. SDM. High levels of calcium signaling proteins, ryanodine receptor 1, RyR1, calsequestrin 1/2, CASQ1/2, annexin A2, ANXA2, and sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA1) pump, ATP2A, were signatures of SDM, and decreased levels of oxidative stress peroxiredoxin 1, PRDX1, thioredoxin-dependent peroxide reductase, PRDX3, or superoxide dismutase [Mn] 2, SOD2, signatures of LDM postflight. Results help to better understand the spatiotemporal molecular adaptation of skeletal muscle and provide a large scale database of skeletal muscle from human spaceflight for the better design of effective CM protocols in future human deep space exploration.
Collapse
Affiliation(s)
- Dieter Blottner
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
- NeuroMuscular System & Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-528-347
| | - Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Gabor Trautmann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Maria Hastermann
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
- NeuroMuscular System & Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | | | - Katharina Block
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Joern Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany
- Department of Pediatrics and Adolescence Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Ulrich Limper
- Department of Anaesthesiology and Intensive Care Medicine, Merheim Medical Center, Witten/Herdecke University, 51109 Cologne, Germany
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
- IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy
| | - Michele Salanova
- Institute of Integrative Neuroanatomy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
- NeuroMuscular System & Signaling Group, Center of Space Medicine and Extreme Environments, 10115 Berlin, Germany
| |
Collapse
|
46
|
Simulated microgravity affects stroma-dependent ex vivo myelopoiesis. Tissue Cell 2023; 80:101987. [PMID: 36481580 DOI: 10.1016/j.tice.2022.101987] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
Microgravity is known negatively affect physiology of living beings, including hematopoiesis. Dysregulation of hematopoietic cells and supporting stroma relationships in bone marrow niche may be in charge. We compared the efficacy of ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs) in presence of native or osteocommitted MSCs under simulated microgravity (Smg) using Random Positioning Machine (RPM). In comparison with 1 g, a decrease of MSC-associated HSPCs and an increase of floating HSPCs was observed after 7 days of Smg exposure. Among floating HSPCs, primitive progenitors were presented by late CD34+/133-. Total CFUs as well as erythroid (BFU-E) and granulocytic (CFU-G) numbers were lower. MSC-associated primitive HSPCs demonstrated increased proportion of late CD34+/133- in expense of early CD34-/133+. Osteo-MSCs preferentially supported late primitive CD34+ and more committed HSPCs as followed from increase of CFUs, and CD235a+ erythroid progenitors. Under Smg, an increased VEGF, eotaxin, and GRO-a levels, and a decrease in RANTES were found in the osteo-MSC-HSPC co-cultures. IL-6,-8, -13, G-CSF, GRO-a, MCP-3, MIP-1b, VEGF increased in co-culture with osteo-MSCs vs intact MSCs. Based on the findings, the misbalance between primitive/committed HSPCs and a decrease in hematopoiesis-supportive activity of osteocommitted cells are supposed to underline hematopoietic disorders during space flights.
Collapse
|
47
|
Cahill T, da Silveira WA, Renaud L, Wang H, Williamson T, Chung D, Chan S, Overton I, Hardiman G. Investigating the effects of chronic low-dose radiation exposure in the liver of a hypothermic zebrafish model. Sci Rep 2023; 13:918. [PMID: 36650199 PMCID: PMC9845366 DOI: 10.1038/s41598-022-26976-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Mankind's quest for a manned mission to Mars is placing increased emphasis on the development of innovative radio-protective countermeasures for long-term space travel. Hibernation confers radio-protective effects in hibernating animals, and this has led to the investigation of synthetic torpor to mitigate the deleterious effects of chronic low-dose-rate radiation exposure. Here we describe an induced torpor model we developed using the zebrafish. We explored the effects of radiation exposure on this model with a focus on the liver. Transcriptomic and behavioural analyses were performed. Radiation exposure resulted in transcriptomic perturbations in lipid metabolism and absorption, wound healing, immune response, and fibrogenic pathways. Induced torpor reduced metabolism and increased pro-survival, anti-apoptotic, and DNA repair pathways. Coupled with radiation exposure, induced torpor led to a stress response but also revealed maintenance of DNA repair mechanisms, pro-survival and anti-apoptotic signals. To further characterise our model of induced torpor, the zebrafish model was compared with hepatic transcriptomic data from hibernating grizzly bears (Ursus arctos horribilis) and active controls revealing conserved responses in gene expression associated with anti-apoptotic processes, DNA damage repair, cell survival, proliferation, and antioxidant response. Similarly, the radiation group was compared with space-flown mice revealing shared changes in lipid metabolism.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Willian Abraham da Silveira
- School of Health, Science and Wellbeing, Department of Biological Sciences, Science Centre, Staffordshire University, Leek Road, Stoke-On-Trent, ST4 2DF, UK
- International Space University, 1 Rue Jean-Dominique Cassini, 67400, Illkirch-Graffenstaden, France
| | - Ludivine Renaud
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hao Wang
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK
| | - Tucker Williamson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Sherine Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
- JLABS at the Children's National Research and Innovation Campus, Washington, DC, 20012, USA
| | - Ian Overton
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, Belfast, BT9 5DL, UK.
- Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
48
|
Anupom T, Vanapalli SA. A Compact Imaging Platform for Conducting C. elegans Phenotypic Assays on Earth and in Spaceflight. Life (Basel) 2023; 13:200. [PMID: 36676149 PMCID: PMC9862956 DOI: 10.3390/life13010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/11/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The model organism Caenorhabditis elegans is used in a variety of applications ranging from fundamental biological studies, to drug screening, to disease modeling, and to space-biology investigations. These applications rely on conducting whole-organism phenotypic assays involving animal behavior and locomotion. In this study, we report a 3D printed compact imaging platform (CIP) that is integrated with a smart-device camera for the whole-organism phenotyping of C. elegans. The CIP has no external optical elements and does not require mechanical focusing, simplifying the optical configuration. The small footprint of the system powered with a standard USB provides capabilities ranging from plug-and-play, to parallel operation, and to housing it in incubators for temperature control. We demonstrate on Earth the compatibility of the CIP with different C. elegans substrates, including agar plates, liquid droplets on glass slides and microfluidic chips. We validate the system with behavioral and thrashing assays and show that the phenotypic readouts are in good agreement with the literature data. We conduct a pilot study with mutants and show that the phenotypic data collected from the CIP distinguishes these mutants. Finally, we discuss how the simplicity and versatility offered by CIP makes it amenable to future C. elegans investigations on the International Space Station, where science experiments are constrained by system size, payload weight and crew time. Overall, the compactness, portability and ease-of-use makes the CIP desirable for research and educational outreach applications on Earth and in space.
Collapse
Affiliation(s)
- Taslim Anupom
- Electrical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | | |
Collapse
|
49
|
Herssens N, Cowburn J, Albracht K, Braunstein B, Cazzola D, Colyer S, Minetti AE, Pavei G, Rittweger J, Weber T, Green DA. Movement in low gravity environments (MoLo) programme-The MoLo-L.O.O.P. study protocol. PLoS One 2022; 17:e0278051. [PMID: 36417480 PMCID: PMC9683620 DOI: 10.1371/journal.pone.0278051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Exposure to prolonged periods in microgravity is associated with deconditioning of the musculoskeletal system due to chronic changes in mechanical stimulation. Given astronauts will operate on the Lunar surface for extended periods of time, it is critical to quantify both external (e.g., ground reaction forces) and internal (e.g., joint reaction forces) loads of relevant movements performed during Lunar missions. Such knowledge is key to predict musculoskeletal deconditioning and determine appropriate exercise countermeasures associated with extended exposure to hypogravity. OBJECTIVES The aim of this paper is to define an experimental protocol and methodology suitable to estimate in high-fidelity hypogravity conditions the lower limb internal joint reaction forces. State-of-the-art movement kinetics, kinematics, muscle activation and muscle-tendon unit behaviour during locomotor and plyometric movements will be collected and used as inputs (Objective 1), with musculoskeletal modelling and an optimisation framework used to estimate lower limb internal joint loading (Objective 2). METHODS Twenty-six healthy participants will be recruited for this cross-sectional study. Participants will walk, skip and run, at speeds ranging between 0.56-3.6 m/s, and perform plyometric movement trials at each gravity level (1, 0.7, 0.5, 0.38, 0.27 and 0.16g) in a randomized order. Through the collection of state-of-the-art kinetics, kinematics, muscle activation and muscle-tendon behaviour, a musculoskeletal modelling framework will be used to estimate lower limb joint reaction forces via tracking simulations. CONCLUSION The results of this study will provide first estimations of internal musculoskeletal loads associated with human movement performed in a range of hypogravity levels. Thus, our unique data will be a key step towards modelling the musculoskeletal deconditioning associated with long term habitation on the Lunar surface, and thereby aiding the design of Lunar exercise countermeasures and mitigation strategies.
Collapse
Affiliation(s)
- Nolan Herssens
- Space Medicine Team, European Astronaut Centre, European Space Agency, Cologne, Germany
| | - James Cowburn
- Department for Health, University of Bath, Bath, United Kingdom
- Centre for the Analysis of Motion, Entertainment Research and Applications, University of Bath, Bath, United Kingdom
| | - Kirsten Albracht
- Centre for Health and Integrative Physiology in Space, German Sport University, Cologne, Germany
- Institute of Movement and Neuroscience, German Sport University, Cologne, Germany
- Department of Medical Engineering and Technomathematics, University of Applied Sciences Aachen, Aachen, Germany
| | - Bjoern Braunstein
- Centre for Health and Integrative Physiology in Space, German Sport University, Cologne, Germany
- Institute of Movement and Neuroscience, German Sport University, Cologne, Germany
- Institute of Biomechanics and Orthopaedics, German Sport University, Cologne, Germany
- German Research Centre of Elite Sport Cologne, Cologne, Germany
| | - Dario Cazzola
- Department for Health, University of Bath, Bath, United Kingdom
- Centre for the Analysis of Motion, Entertainment Research and Applications, University of Bath, Bath, United Kingdom
| | - Steffi Colyer
- Department for Health, University of Bath, Bath, United Kingdom
- Centre for the Analysis of Motion, Entertainment Research and Applications, University of Bath, Bath, United Kingdom
| | - Alberto E. Minetti
- Laboratory of Physiomechanics of Locomotion, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gaspare Pavei
- Laboratory of Physiomechanics of Locomotion, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Jörn Rittweger
- Division of Muscle and Bone Metabolism, Institute of Aerospace Medicine DLR, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Tobias Weber
- Space Medicine Team, European Astronaut Centre, European Space Agency, Cologne, Germany
- KBR, Cologne, North Rhein-Westphalia, Germany
| | - David A. Green
- Space Medicine Team, European Astronaut Centre, European Space Agency, Cologne, Germany
- KBR, Cologne, North Rhein-Westphalia, Germany
- Centre of Human and Applied Physiological Sciences, King’s College London, London, United Kingdom
| |
Collapse
|
50
|
Changes in interstitial fluid flow, mass transport and the bone cell response in microgravity and normogravity. Bone Res 2022; 10:65. [PMID: 36411278 PMCID: PMC9678891 DOI: 10.1038/s41413-022-00234-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, our scientific interest in spaceflight has grown exponentially and resulted in a thriving area of research, with hundreds of astronauts spending months of their time in space. A recent shift toward pursuing territories farther afield, aiming at near-Earth asteroids, the Moon, and Mars combined with the anticipated availability of commercial flights to space in the near future, warrants continued understanding of the human physiological processes and response mechanisms when in this extreme environment. Acute skeletal loss, more severe than any bone loss seen on Earth, has significant implications for deep space exploration, and it remains elusive as to why there is such a magnitude of difference between bone loss on Earth and loss in microgravity. The removal of gravity eliminates a critical primary mechano-stimulus, and when combined with exposure to both galactic and solar cosmic radiation, healthy human tissue function can be negatively affected. An additional effect found in microgravity, and one with limited insight, involves changes in dynamic fluid flow. Fluids provide the most fundamental way to transport chemical and biochemical elements within our bodies and apply an essential mechano-stimulus to cells. Furthermore, the cell cytoplasm is not a simple liquid, and fluid transport phenomena together with viscoelastic deformation of the cytoskeleton play key roles in cell function. In microgravity, flow behavior changes drastically, and the impact on cells within the porous system of bone and the influence of an expanding level of adiposity are not well understood. This review explores the role of interstitial fluid motion and solute transport in porous bone under two different conditions: normogravity and microgravity.
Collapse
|