1
|
Forghani P, Liu W, Wang Z, Ling Z, Takaesu F, Yang E, Tharp GK, Nielsen S, Doraisingam S, Countryman S, Davis ME, Wu R, Jia S, Xu C. Spaceflight alters protein levels and gene expression associated with stress response and metabolic characteristics in human cardiac spheroids. Biomaterials 2025; 317:123080. [PMID: 39809079 PMCID: PMC11788069 DOI: 10.1016/j.biomaterials.2024.123080] [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: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) possess tremendous advantage for cardiac regeneration. However, cell survival is challenging upon cell transplantation. Since microgravity can profoundly affect cellular properties, we investigated the effect of spaceflight on hiPSC-CMs. Cardiac spheroids derived from hiPSCs were transported to the International Space Station (ISS) via the SpaceX Crew-8 mission and cultured under space microgravity for 8 days. Beating cardiac spheroids were observed on the ISS and upon successful experimentation by the astronauts in space, the live cultures were returned to Earth. These cells had normal displacement (an indicator of contraction) and Ca2+ transient parameters in 3D live cell imaging. Proteomic analysis revealed that spaceflight upregulated many proteins involved in metabolism (n = 90), cellular component of mitochondrion (n = 62) and regulation of proliferation (n = 10). Specific metabolic pathways enriched by spaceflight included glutathione metabolism, biosynthesis of amino acids, and pyruvate metabolism. In addition, the top upregulated proteins in spaceflight samples included those involved in cellular stress response, cell survival, and metabolism. Transcriptomic profiles indicated that spaceflight upregulated genes associated with cardiomyocyte development, and cellular components of cardiac structure and mitochondrion. Furthermore, spaceflight upregulated genes in metabolic pathways associated with cell survival such as glycerophospholipid metabolism and glycerolipid metabolism. These findings indicate that short-term exposure of 3D hiPSC-CMs to the space environment led to significant changes in protein levels and gene expression involved in cell survival and metabolism.
Collapse
Affiliation(s)
- Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zeyu Wang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhi Ling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Felipe Takaesu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Evan Yang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Gregory K Tharp
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | | | | | - Michael E Davis
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
2
|
Kahlert S, Nossol C, Krüger M, Kopp S, Grimm D, Wuest SL, Rothkötter HJ. Dynamic Mechanical Load as a Trigger for Growth and Proliferation in Porcine Epithelial Cells. Biomolecules 2025; 15:455. [PMID: 40149991 PMCID: PMC11940287 DOI: 10.3390/biom15030455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The impact of gravity is a basic force determining our existence on Earth. Changes in orientation with respect to the gravity vector trigger alternating mechanical forces on organisms, organs, and cells. In the intestines of mammals, epithelial cells are continuously exposed to changed orientations to gravity. In this study, we employed dynamic cultivation systems to mimic the load changes and the resulting mechanical forces. The morphological and functional response of non-cancer-derived porcine epithelial cell lines IPEC-1 and IPEC-J2 was analyzed. We found that dynamic growth conditions affect morphology in the enterocyte model IPEC-1 but not in IPEC-J2. Changes in IPEC-1 were accompanied by modifications of the distribution and structure of the F-actin cytoskeleton rather than the amount. The structure of the apical brush border and the tight junction system seemed to be largely unaffected; however, a robust decrease in transepithelial resistance was found in IPEC-1 and partially in IPEC-J2. We further detected an increase in Ki67, pointing towards accelerated proliferation. In line with this finding, we detected a doubling of cellular mitochondrial respiration, which was not linked to a general increase in the respiratory chain capacity. Dynamic cultivation of confluent epithelial cell layers did not evoke signs of senescence. In summary, we identified the mechanical load cycle as a relevant parameter for the modulation of the morphological structure and physiological behaviour of intestinal epithelial cells.
Collapse
Affiliation(s)
- Stefan Kahlert
- Institut für Anatomie, Medizinische Fakultät, Otto von Guericke Universität Magdeburg, Leipziger Str. 44, Haus 43, 39120 Magdeburg, Germany; (C.N.); (H.-J.R.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke Universität Magdeburg, 39106 Magdeburg, Germany; (M.K.); (S.K.); (D.G.)
| | - Constanze Nossol
- Institut für Anatomie, Medizinische Fakultät, Otto von Guericke Universität Magdeburg, Leipziger Str. 44, Haus 43, 39120 Magdeburg, Germany; (C.N.); (H.-J.R.)
| | - Marcus Krüger
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke Universität Magdeburg, 39106 Magdeburg, Germany; (M.K.); (S.K.); (D.G.)
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Sascha Kopp
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke Universität Magdeburg, 39106 Magdeburg, Germany; (M.K.); (S.K.); (D.G.)
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
- Core Facility Tissue Engineering, Institut für Chemie, Otto von Guericke Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke Universität Magdeburg, 39106 Magdeburg, Germany; (M.K.); (S.K.); (D.G.)
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, HØegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
| | - Simon L. Wuest
- Space Biology Group, Institute of Medical Engineering, Lucerne, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, Obermattweg 9, 6052 Hergiswil, NW, Switzerland
| | - Hermann-Josef Rothkötter
- Institut für Anatomie, Medizinische Fakultät, Otto von Guericke Universität Magdeburg, Leipziger Str. 44, Haus 43, 39120 Magdeburg, Germany; (C.N.); (H.-J.R.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke Universität Magdeburg, 39106 Magdeburg, Germany; (M.K.); (S.K.); (D.G.)
| |
Collapse
|
3
|
Paredes-Espinosa MB, Paluh JL. Synthetic embryology of the human heart. Front Cell Dev Biol 2025; 12:1478549. [PMID: 39935786 PMCID: PMC11810959 DOI: 10.3389/fcell.2024.1478549] [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: 08/10/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
The evolution of stem cell-based heart models from cells and tissues to organoids and assembloids and recently synthetic embryology gastruloids, is poised to revolutionize our understanding of cardiac development, congenital to adult diseases, and patient customized therapies. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have already been integrated into transplantable patches and are in preclinical efforts to reverse fibrotic scarring from myocardial infarctions. To inform on the complexity of heart diseases, multi-tissue morphogenic heart models are needed that replicate fundamental components of heart function to heart organogenesis in vitro and which require a deep understanding of heart development. Organoid and assembloid models capture selected multicellular cardiac processes, such as chamber formation and priming events for vascularization. Gastruloid heart models offer deeper insights as synthetic embryology to mimic multi-staged developmental events of in vivo heart organogenesis including established heart fields, crescent formation and heart tube development along with vascular systemic foundation and even further steps. The human Elongating Multi-Lineage Organized Cardiac (EMLOC) gastruloid model captures these stages and additional events including chamber genesis, patterned vascularization, and extrinsic central and intrinsic cardiac nervous system (CNS-ICNS) integration guided by spatiotemporal and morphogenic processes with neural crest cells. Gastruloid synthetic embryology heart models offer new insights into previously hidden processes of development and provide powerful platforms for addressing heart disease that extends beyond cardiomyocytes, such as arrhythmogenic diseases, congenital defects, and systemic injury interactions, as in spinal cord injuries. The holistic view that is emerging will reveal heart development and disease in unprecedented detail to drive transformative state-of-the-art innovative applications for heart health.
Collapse
Affiliation(s)
| | - Janet L. Paluh
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science and Engineering, University at Albany, Albany, NY, United States
| |
Collapse
|
4
|
Marotta D, Ijaz L, Barbar L, Nijsure M, Stein J, Pirjanian N, Kruglikov I, Clements T, Stoudemire J, Grisanti P, Noggle SA, Loring JF, Fossati V. Effects of microgravity on human iPSC-derived neural organoids on the International Space Station. Stem Cells Transl Med 2024; 13:1186-1197. [PMID: 39441987 PMCID: PMC11631337 DOI: 10.1093/stcltm/szae070] [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: 03/06/2024] [Accepted: 07/30/2024] [Indexed: 10/25/2024] Open
Abstract
Research conducted on the International Space Station (ISS) in low-Earth orbit (LEO) has shown the effects of microgravity on multiple organs. To investigate the effects of microgravity on the central nervous system, we developed a unique organoid strategy for modeling specific regions of the brain that are affected by neurodegenerative diseases. We generated 3-dimensional human neural organoids from induced pluripotent stem cells (iPSCs) derived from individuals affected by primary progressive multiple sclerosis (PPMS) or Parkinson's disease (PD) and non-symptomatic controls, by differentiating them toward cortical and dopaminergic fates, respectively, and combined them with isogenic microglia. The organoids were cultured for a month using a novel sealed cryovial culture method on the International Space Station (ISS) and a parallel set that remained on Earth. Live samples were returned to Earth for analysis by RNA expression and histology and were attached to culture dishes to enable neurite outgrowth. Our results show that both cortical and dopaminergic organoids cultured in LEO had lower levels of genes associated with cell proliferation and higher levels of maturation-associated genes, suggesting that the cells matured more quickly in LEO. This study is continuing with several more missions in order to understand the mechanisms underlying accelerated maturation and to investigate other neurological diseases. Our goal is to make use of the opportunity to study neural cells in LEO to better understand and treat neurodegenerative disease on Earth and to help ameliorate potentially adverse neurological effects of space travel.
Collapse
Affiliation(s)
- Davide Marotta
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Laraib Ijaz
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Lilianne Barbar
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Madhura Nijsure
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Jason Stein
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, United States
| | - Nicolette Pirjanian
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Ilya Kruglikov
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | | | | | - Paula Grisanti
- National Stem Cell Foundation, Louisville, KY 40202, United States
| | - Scott A Noggle
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| | - Jeanne F Loring
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, United States
- National Stem Cell Foundation, Louisville, KY 40202, United States
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, United States
| |
Collapse
|
5
|
Blazeski A, Garcia-Cardena G, Kamm RD. Advancing Cardiac Organoid Engineering Through Application of Biophysical Forces. IEEE Rev Biomed Eng 2024; PP:211-230. [PMID: 40030454 DOI: 10.1109/rbme.2024.3514378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cardiac organoids represent an important bioengineering opportunity in the development of models to study human heart pathophysiology. By incorporating multiple cardiac cell types in three-dimensional culture and developmentally-guided biochemical signaling, cardiac organoids recapitulate numerous features of heart tissue. However, cardiac tissue also experiences a variety of mechanical forces as the heart develops and over the course of each contraction cycle. It is now clear that these forces impact cellular specification, phenotype, and function, and should be incorporated into the engineering of cardiac organoids in order to generate better models. In this review, we discuss strategies for engineering cardiac organoids and report the effects of organoid design on the function of cardiac cells. We then discuss the mechanical environment of the heart, including forces arising from tissue elasticity, contraction, blood flow, and stretch, and report on efforts to mimic these biophysical cues in cardiac organoids. Finally, we review emerging areas of cardiac organoid research, for the study of cardiac development, the formation of multi-organ models, and the simulation of the effects of spaceflight on cardiac tissue and consider how these investigations might benefit from the inclusion of mechanical cues.
Collapse
|
6
|
Harriot AD, Ward CW, Kim DH. Microphysiological systems to advance human pathophysiology and translational medicine. J Appl Physiol (1985) 2024; 137:1494-1501. [PMID: 39417817 DOI: 10.1152/japplphysiol.00087.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Microphysiological systems (MPS) or "organ-on-a-chip" models are sophisticated tools that harness techniques from cell biology, tissue engineering, and microengineering to recapitulate human physiology. Typically, MPS are biofabricated three-dimensional (3-D) tissue constructs integrated into platforms designed to mimic the tissue microenvironment and provide functional outputs. Over the past decade, researchers have endeavored to manufacture high-throughput, high-fidelity MPS models of all major human organs. By incorporating patient-derived cells, researchers have produced biomimetic models of tissues with disease-linked genetic mutations capable of exhibiting patient heterogeneity. This work has demonstrated that MPS more closely model organotypic function and pathophysiology than traditional two-dimensional (2-D) culture systems. Moreover, investigators have shown that human MPS are better predictors of drug efficacy and toxicity than animal models. Thus, MPS have emerged as a promising candidate to improve the efficacy and safety of preclinical trials. In this mini-review, we provide an overview of current advances in MPS models, their applications in mechanistic research, and relevance to drug screening. Finally, we discuss current investments in MPS development by the United States federal government and research institutions around the world to advance translational medicine.
Collapse
Affiliation(s)
- Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Division of Geriatric Medicine and Gerontology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher W Ward
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Claude D. Pepper Older Americans Independence Center, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, Maryland, United States
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
7
|
Mozneb M, Arzt M, Mesci P, Martin DMN, Pohlman S, Lawless G, Doraisingam S, Al Neyadi S, Barnawi R, Al Qarni A, Whitson PA, Shoffner J, Stoudemire J, Countryman S, Svendsen CN, Sharma A. Surface tension enables induced pluripotent stem cell culture in commercially available hardware during spaceflight. NPJ Microgravity 2024; 10:97. [PMID: 39402072 PMCID: PMC11473755 DOI: 10.1038/s41526-024-00435-y] [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: 02/13/2024] [Accepted: 10/06/2024] [Indexed: 10/17/2024] Open
Abstract
Low Earth Orbit (LEO) has emerged as a unique environment for evaluating altered stem cell properties in microgravity. LEO has become increasingly accessible for research and development due to progress in private spaceflight. Axiom Mission 2 (Ax-2) was launched as the second all-private astronaut mission to the International Space Station (ISS). Frozen human induced pluripotent stem cells (hiPSCs) expressing green fluorescent protein (GFP) under the SOX2 promoter, as well as fibroblasts differentiated from SOX2-GFP hiPSCs, were sent to the ISS. Astronauts then thawed and seeded both cell types into commercially available 96-well plates, which provided surface tension that reduced fluid movement out of individual wells and showed that hiPSCs or hiPSC-derived fibroblasts could survive either in suspension or attached to a Matrigel substrate. Furthermore, both cell types could be transfected with red fluorescent protein (RFP)-expressing plasmid. We demonstrate that hiPSCs and hiPSC-fibroblasts can be thawed in microgravity in off-the-shelf, commercially-available cell culture hardware, can associate into 3D spheroids or grow adherently in Matrigel, and can be transfected with DNA. This lays the groundwork for future biomanufacturing experiments in space.
Collapse
Affiliation(s)
- Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - George Lawless
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Rayyanah Barnawi
- Axiom Space, Inc., Houston, TX, USA
- Saudi Space Commission, Riyadh, Saudi Arabia
| | - Ali Al Qarni
- Axiom Space, Inc., Houston, TX, USA
- Saudi Space Commission, Riyadh, Saudi Arabia
| | | | | | | | | | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
8
|
McCain ML. Heart-on-a-Chip at the final frontier. Proc Natl Acad Sci U S A 2024; 121:e2417412121. [PMID: 39348548 PMCID: PMC11474057 DOI: 10.1073/pnas.2417412121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024] Open
Affiliation(s)
- Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
9
|
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
|
10
|
Han H, Jia H, Wang YF, Song JP. Cardiovascular adaptations and pathological changes induced by spaceflight: from cellular mechanisms to organ-level impacts. Mil Med Res 2024; 11:68. [PMID: 39334239 PMCID: PMC11429428 DOI: 10.1186/s40779-024-00570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
The advancement in extraterrestrial exploration has highlighted the crucial need for studying how the human cardiovascular system adapts to space conditions. Human development occurs under the influence of gravity, shielded from space radiation by Earth's magnetic field, and within an environment characterized by 24-hour day-night cycles resulting from Earth's rotation, thus deviating from these conditions necessitates adaptive responses for survival. With upcoming manned lunar and Martian missions approaching rapidly, it is essential to understand the impact of various stressors induced by outer-space environments on cardiovascular health. This comprehensive review integrates insights from both actual space missions and simulated experiments on Earth, to analyze how microgravity, space radiation, and disrupted circadian affect cardiovascular well-being. Prolonged exposure to microgravity induces myocardial atrophy and endothelial dysfunction, which may be exacerbated by space radiation. Mitochondrial dysfunction and oxidative stress emerge as key underlying mechanisms along with disturbances in ion channel perturbations, cytoskeletal damage, and myofibril changes. Disruptions in circadian rhythms caused by factors such as microgravity, light exposure, and irregular work schedules, could further exacerbate cardiovascular issues. However, current research tends to predominantly focus on disruptions in the core clock gene, overlooking the multifactorial nature of circadian rhythm disturbances in space. Future space missions should prioritize targeted prevention strategies and early detection methods for identifying cardiovascular risks, to preserve astronaut health and ensure mission success.
Collapse
Affiliation(s)
- Han Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yi-Fan Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiang-Ping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
11
|
Abdelfattah F, Schulz H, Wehland M, Corydon TJ, Sahana J, Kraus A, Krüger M, González-Torres LF, Cortés-Sánchez JL, Wise PM, Mushunuri A, Hemmersbach R, Liemersdorf C, Infanger M, Grimm D. Omics Studies of Specialized Cells and Stem Cells under Microgravity Conditions. Int J Mol Sci 2024; 25:10014. [PMID: 39337501 PMCID: PMC11431953 DOI: 10.3390/ijms251810014] [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: 08/20/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
The primary objective of omics in space with focus on the human organism is to characterize and quantify biological factors that alter structure, morphology, function, and dynamics of human cells exposed to microgravity. This review discusses exciting data regarding genomics, transcriptomics, epigenomics, metabolomics, and proteomics of human cells and individuals in space, as well as cells cultured under simulated microgravity. The NASA Twins Study significantly heightened interest in applying omics technologies and bioinformatics in space and terrestrial environments. Here, we present the available publications in this field with a focus on specialized cells and stem cells exposed to real and simulated microgravity conditions. We summarize current knowledge of the following topics: (i) omics studies on stem cells, (ii) omics studies on benign specialized different cell types of the human organism, (iii) discussing the advantages of this knowledge for space commercialization and exploration, and (iv) summarizing the emerging opportunities for translational regenerative medicine for space travelers and human patients on Earth.
Collapse
Affiliation(s)
- Fatima Abdelfattah
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| | - Armin Kraus
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Luis Fernando González-Torres
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - José Luis Cortés-Sánchez
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Ashwini Mushunuri
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
| | - Ruth Hemmersbach
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Christian Liemersdorf
- Department of Applied Aerospace Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (R.H.); (C.L.)
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (F.A.); (H.S.); (M.W.); (A.K.); (M.K.); (L.F.G.-T.); (J.L.C.-S.); (P.M.W.); (A.M.); (M.I.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (T.J.C.); (J.S.)
| |
Collapse
|
12
|
Hariom SK, Nelson EJR. Cardiovascular adaptations in microgravity conditions. LIFE SCIENCES IN SPACE RESEARCH 2024; 42:64-71. [PMID: 39067992 DOI: 10.1016/j.lssr.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/30/2024]
Abstract
Gravity has had a significant impact on the evolution of life on Earth with organisms developing necessary biological adaptations over billions of years to counter this ever-existing force. There has been an exponential increase in experiments using real and simulated gravity environments in the recent years. Although an understanding followed by discovery of counter measures to negate diminished gravity in space had been the driving force of research initially, there has since been a phenomenal leap wherein a force unearthly as microgravity is beginning to show promising potential. The current review summarizes pathophysiological changes that occur in multiple aspects of the cardiovascular system when exposed to an altered gravity environment leading to cardiovascular deconditioning and orthostatic intolerance. Gravity influences not just the complex multicellular systems but even the survival of organisms at the molecular level by intervening fundamental cellular processes, directly affecting those linked to actin and microtubule organization via mechano-transduction pathways. The reach of gravity ranges from cytoskeletal rearrangement that regulates cell adhesion and migration to intracellular dynamics that dictate cell fate commitment and differentiation. An understanding that microgravity itself is not present on Earth propels the scope of simulated gravity conditions to be a unique and useful environment that could be explored for enhancing the potential of stem cells for a wide range of applications as has been highlighted here.
Collapse
Affiliation(s)
- Senthil Kumar Hariom
- Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, TN, India
| | - Everette Jacob Remington Nelson
- Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632 014, TN, India.
| |
Collapse
|
13
|
Kim J, Tierney BT, Overbey EG, Dantas E, Fuentealba M, Park J, Narayanan SA, Wu F, Najjar D, Chin CR, Meydan C, Loy C, Mathyk B, Klotz R, Ortiz V, Nguyen K, Ryon KA, Damle N, Houerbi N, Patras LI, Schanzer N, Hutchinson GA, Foox J, Bhattacharya C, Mackay M, Afshin EE, Hirschberg JW, Kleinman AS, Schmidt JC, Schmidt CM, Schmidt MA, Beheshti A, Matei I, Lyden D, Mullane S, Asadi A, Lenz JS, Mzava O, Yu M, Ganesan S, De Vlaminck I, Melnick AM, Barisic D, Winer DA, Zwart SR, Crucian BE, Smith SM, Mateus J, Furman D, Mason CE. Single-cell multi-ome and immune profiles of the Inspiration4 crew reveal conserved, cell-type, and sex-specific responses to spaceflight. Nat Commun 2024; 15:4954. [PMID: 38862516 PMCID: PMC11166952 DOI: 10.1038/s41467-024-49211-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Spaceflight induces an immune response in astronauts. To better characterize this effect, we generated single-cell, multi-ome, cell-free RNA (cfRNA), biochemical, and hematology data for the SpaceX Inspiration4 (I4) mission crew. We found that 18 cytokines/chemokines related to inflammation, aging, and muscle homeostasis changed after spaceflight. In I4 single-cell multi-omics data, we identified a "spaceflight signature" of gene expression characterized by enrichment in oxidative phosphorylation, UV response, immune function, and TCF21 pathways. We confirmed the presence of this signature in independent datasets, including the NASA Twins Study, the I4 skin spatial transcriptomics, and 817 NASA GeneLab mouse transcriptomes. Finally, we observed that (1) T cells showed an up-regulation of FOXP3, (2) MHC class I genes exhibited long-term suppression, and (3) infection-related immune pathways were associated with microbiome shifts. In summary, this study reveals conserved and distinct immune disruptions occurring and details a roadmap for potential countermeasures to preserve astronaut health.
Collapse
Affiliation(s)
- JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Braden T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Center for STEM, University of Austin, Austin, TX, USA
- BioAstra, Inc, New York, NY, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Matias Fuentealba
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Jiwoon Park
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - S Anand Narayanan
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Fei Wu
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Christopher R Chin
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Conor Loy
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Begum Mathyk
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Khiem Nguyen
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Namita Damle
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
| | - Nadia Houerbi
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laura I Patras
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Nathan Schanzer
- School of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Gwyneth A Hutchinson
- NASA Center for the Utilization of Biological Engineering in Space (CUBES), Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Chandrima Bhattacharya
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Matthew Mackay
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Evan E Afshin
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jeremy Wain Hirschberg
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Julian C Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
| | - Caleb M Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
- Department of Systems Engineering, Colorado State University, Fort Collins, CO, USA
| | - Michael A Schmidt
- Sovaris Aerospace, Boulder, CO, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO, USA
| | - Afshin Beheshti
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Irina Matei
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - David Lyden
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Sean Mullane
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Amran Asadi
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Joan S Lenz
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Omary Mzava
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Saravanan Ganesan
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Iwijn De Vlaminck
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, 14850, USA
| | - Ari M Melnick
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Darko Barisic
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Daniel A Winer
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Division of Cellular & Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sara R Zwart
- University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA
| | - Brian E Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Human Health and Performance Directorate, 2101 NASA Parkway, Houston, TX, 77058, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Human Health and Performance Directorate, 2101 NASA Parkway, Houston, TX, 77058, USA
| | - Jaime Mateus
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - David Furman
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Stanford 1000 Immunomes Project, Stanford School of Medicine, Stanford, CA, 94306, USA.
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, CONICET, Pilar, Argentina.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 100221, USA.
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA.
- Tri-Institutional Biology and Medicine Program, Weill Cornell Medicine, New York, NY, 10021, USA.
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
14
|
Chua CYX, Jimenez M, Mozneb M, Traverso G, Lugo R, Sharma A, Svendsen CN, Wagner WR, Langer R, Grattoni A. Advanced material technologies for space and terrestrial medicine. NATURE REVIEWS MATERIALS 2024; 9:808-821. [DOI: 10.1038/s41578-024-00691-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 01/05/2025]
|
15
|
Han X, Qu L, Yu M, Ye L, Shi L, Ye G, Yang J, Wang Y, Fan H, Wang Y, Tan Y, Wang C, Li Q, Lei W, Chen J, Liu Z, Shen Z, Li Y, Hu S. Thiamine-modified metabolic reprogramming of human pluripotent stem cell-derived cardiomyocyte under space microgravity. Signal Transduct Target Ther 2024; 9:86. [PMID: 38584163 PMCID: PMC10999445 DOI: 10.1038/s41392-024-01791-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 02/08/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
During spaceflight, the cardiovascular system undergoes remarkable adaptation to microgravity and faces the risk of cardiac remodeling. Therefore, the effects and mechanisms of microgravity on cardiac morphology, physiology, metabolism, and cellular biology need to be further investigated. Since China started constructing the China Space Station (CSS) in 2021, we have taken advantage of the Shenzhou-13 capsule to send human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) to the Tianhe core module of the CSS. In this study, hPSC-CMs subjected to space microgravity showed decreased beating rate and abnormal intracellular calcium cycling. Metabolomic and transcriptomic analyses revealed a battery of metabolic remodeling of hPSC-CMs in spaceflight, especially thiamine metabolism. The microgravity condition blocked the thiamine intake in hPSC-CMs. The decline of thiamine utilization under microgravity or by its antagonistic analog amprolium affected the process of the tricarboxylic acid cycle. It decreased ATP production, which led to cytoskeletal remodeling and calcium homeostasis imbalance in hPSC-CMs. More importantly, in vitro and in vivo studies suggest that thiamine supplementation could reverse the adaptive changes induced by simulated microgravity. This study represents the first astrobiological study on the China Space Station and lays a solid foundation for further aerospace biomedical research. These data indicate that intervention of thiamine-modified metabolic reprogramming in human cardiomyocytes during spaceflight might be a feasible countermeasure against microgravity.
Collapse
Affiliation(s)
- Xinglong Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Lina Qu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Lingqun Ye
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Liujia Shi
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Guangfu Ye
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Jingsi Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Hao Fan
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yong Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Qi Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Jianghai Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoxia Liu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China.
| |
Collapse
|
16
|
Garcia-Medina JS, Sienkiewicz K, Narayanan SA, Overbey EG, Grigorev K, Ryon KA, Burke M, Proszynski J, Tierney B, Schmidt CM, Mencia-Trinchant N, Klotz R, Ortiz V, Foox J, Chin C, Najjar D, Matei I, Chan I, Cruchaga C, Kleinman A, Kim J, Lucaci A, Loy C, Mzava O, De Vlaminck I, Singaraju A, Taylor LE, Schmidt JC, Schmidt MA, Blease K, Moreno J, Boddicker A, Zhao J, Lajoie B, Altomare A, Kruglyak S, Levy S, Yu M, Hassane DC, Bailey SM, Bolton K, Mateus J, Mason CE. Genome and clonal hematopoiesis stability contrasts with immune, cfDNA, mitochondrial, and telomere length changes during short duration spaceflight. PRECISION CLINICAL MEDICINE 2024; 7:pbae007. [PMID: 38634106 PMCID: PMC11022651 DOI: 10.1093/pcmedi/pbae007] [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: 01/29/2024] [Accepted: 03/24/2024] [Indexed: 04/19/2024] Open
Abstract
Background The Inspiration4 (I4) mission, the first all-civilian orbital flight mission, investigated the physiological effects of short-duration spaceflight through a multi-omic approach. Despite advances, there remains much to learn about human adaptation to spaceflight's unique challenges, including microgravity, immune system perturbations, and radiation exposure. Methods To provide a detailed genetics analysis of the mission, we collected dried blood spots pre-, during, and post-flight for DNA extraction. Telomere length was measured by quantitative PCR, while whole genome and cfDNA sequencing provided insight into genomic stability and immune adaptations. A robust bioinformatic pipeline was used for data analysis, including variant calling to assess mutational burden. Result Telomere elongation occurred during spaceflight and shortened after return to Earth. Cell-free DNA analysis revealed increased immune cell signatures post-flight. No significant clonal hematopoiesis of indeterminate potential (CHIP) or whole-genome instability was observed. The long-term gene expression changes across immune cells suggested cellular adaptations to the space environment persisting months post-flight. Conclusion Our findings provide valuable insights into the physiological consequences of short-duration spaceflight, with telomere dynamics and immune cell gene expression adapting to spaceflight and persisting after return to Earth. CHIP sequencing data will serve as a reference point for studying the early development of CHIP in astronauts, an understudied phenomenon as previous studies have focused on career astronauts. This study will serve as a reference point for future commercial and non-commercial spaceflight, low Earth orbit (LEO) missions, and deep-space exploration.
Collapse
Affiliation(s)
- J Sebastian Garcia-Medina
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Karolina Sienkiewicz
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - S Anand Narayanan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
- BioAstra Inc, New York, NY, USA
| | - Kirill Grigorev
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Krista A Ryon
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Marissa Burke
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Jacqueline Proszynski
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Braden Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Caleb M Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
- Department of Systems Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Nuria Mencia-Trinchant
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Remi Klotz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Veronica Ortiz
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Christopher Chin
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
- BioAstra Inc, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Deena Najjar
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Irenaeus Chan
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Carlos Cruchaga
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Ashley Kleinman
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alexander Lucaci
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Conor Loy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Omary Mzava
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Anvita Singaraju
- Department of Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Julian C Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
| | - Michael A Schmidt
- Sovaris Aerospace, Boulder, CO 80302, USA
- Advanced Pattern Analysis & Human Performance Group, Boulder, CO 80302, USA
| | | | - Juan Moreno
- Element Biosciences, San Diego, CA 10055, USA
| | | | - Junhua Zhao
- Element Biosciences, San Diego, CA 10055, USA
| | | | | | | | - Shawn Levy
- Element Biosciences, San Diego, CA 10055, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Duane C Hassane
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Kelly Bolton
- Washington University St. Louis Oncology Division, St. Louis, MO 63100, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation, Hawthorne, CA 90250, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
- BioAstra Inc, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA
- WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
17
|
Arzt M, Mozneb M, Escopete S, Moses J, Sharma A. The Benefits of Stem Cell Biology and Tissue Engineering in Low-Earth Orbit. Stem Cells Dev 2024; 33:143-147. [PMID: 38326760 DOI: 10.1089/scd.2023.0291] [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: 02/09/2024] Open
Abstract
Over the past 15 years, there has been a significant shift in biomedical research toward a major focus on stem cell research. Although stem cells and their derivatives exhibit potential in modeling and mitigating human diseases, the ongoing objective is to enhance their utilization and translational potential. Stem cells are increasingly employed in both academic and commercial settings for a variety of in vitro and in vivo applications in regenerative medicine. Notably, accessibility to stem cell research in low-Earth orbit (LEO) has expanded, driven by the unique properties of space, such as microgravity, which cannot exactly be replicated on Earth. As private enterprises continue to grow and launch low-orbit payloads alongside government-funded spaceflight, space has evolved into a more viable destination for scientific exploration. This review underscores the potential benefits of microgravity on fundamental stem cell properties, highlighting the adaptability of cells to their environment and emphasizing physical stimuli as a key factor influencing cultured cells. Previous studies suggest that stimuli such as magnetic fields, shear stress, or gravity impact not only cell kinetics, including differentiation and proliferation, but also therapeutic effects such as cells with improved immunosuppressive capabilities or the ability to identify novel targets to refine disease treatments. With the rapid progress and sustained advocacy for space research, we propose that the advantageous properties of LEO create novel opportunities in biomanufacturing for regenerative medicine, spanning disease modeling, the development of stem cell-derived products, and biofabrication.
Collapse
Affiliation(s)
- Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sean Escopete
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Graduate PhD Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jemima Moses
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- CIRM Bridges to Stem Cell Research Program, California State University, Channel Islands, California, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
18
|
Gao M, Guo H, Dong X, Wang Z, Yang Z, Shang Q, Wang Q. Regulation of inflammation during wound healing: the function of mesenchymal stem cells and strategies for therapeutic enhancement. Front Pharmacol 2024; 15:1345779. [PMID: 38425646 PMCID: PMC10901993 DOI: 10.3389/fphar.2024.1345779] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
A wound takes a long time to heal and involves several steps. Following tissue injury, inflammation is the primary cause of tissue regeneration and repair processes. As a result, the pathophysiological processes involving skin damage, healing, and remodeling depend critically on the control of inflammation. The fact that it is a feasible target for improving the prognosis of wound healing has lately become clear. Mesenchymal stem cells (MSCs) are an innovative and effective therapeutic option for wound healing due to their immunomodulatory and paracrine properties. By controlling the inflammatory milieu of wounds through immunomodulation, transplanted MSCs have been shown to speed up the healing process. In addition to other immunomodulatory mechanisms, including handling neutrophil activity and modifying macrophage polarization, there may be modifications to the activation of T cells, natural killer (NK) cells, and dendritic cells (DCs). Furthermore, several studies have shown that pretreating MSCs improves their ability to modulate immunity. In this review, we summarize the existing knowledge about how MSCs influence local inflammation in wounds by influencing immunity to facilitate the healing process. We also provide an overview of MSCs optimizing techniques when used to treat wounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiying Wang
- Department of Plastic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Tomsia M, Cieśla J, Śmieszek J, Florek S, Macionga A, Michalczyk K, Stygar D. Long-term space missions' effects on the human organism: what we do know and what requires further research. Front Physiol 2024; 15:1284644. [PMID: 38415007 PMCID: PMC10896920 DOI: 10.3389/fphys.2024.1284644] [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: 08/28/2023] [Accepted: 01/22/2024] [Indexed: 02/29/2024] Open
Abstract
Space has always fascinated people. Many years have passed since the first spaceflight, and in addition to the enormous technological progress, the level of understanding of human physiology in space is also increasing. The presented paper aims to summarize the recent research findings on the influence of the space environment (microgravity, pressure differences, cosmic radiation, etc.) on the human body systems during short-term and long-term space missions. The review also presents the biggest challenges and problems that must be solved in order to extend safely the time of human stay in space. In the era of increasing engineering capabilities, plans to colonize other planets, and the growing interest in commercial space flights, the most topical issues of modern medicine seems to be understanding the effects of long-term stay in space, and finding solutions to minimize the harmful effects of the space environment on the human body.
Collapse
Affiliation(s)
- Marcin Tomsia
- Department of Forensic Medicine and Forensic Toxicology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Julia Cieśla
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Joanna Śmieszek
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Szymon Florek
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agata Macionga
- School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
20
|
Lecoq PE, Dupuis C, Mousset X, Benoit-Gonnin X, Peyrin JM, Aider JL. Influence of microgravity on spontaneous calcium activity of primary hippocampal neurons grown in microfluidic chips. NPJ Microgravity 2024; 10:15. [PMID: 38321051 PMCID: PMC10847089 DOI: 10.1038/s41526-024-00355-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
The influence of variations of gravity, either hypergravity or microgravity, on the brain of astronauts is a major concern for long journeys in space, to the Moon or to Mars, or simply long-duration missions on the ISS (International Space Station). Monitoring brain activity, before and after ISS missions already demonstrated important and long term effects on the brains of astronauts. In this study, we focus on the influence of gravity variations at the cellular level on primary hippocampal neurons. A dedicated setup has been designed and built to perform live calcium imaging during parabolic flights. During a CNES (Centre National d'Etudes Spatiales) parabolic flight campaign, we were able to observe and monitor the calcium activity of 2D networks of neurons inside microfluidic devices during gravity changes over different parabolas. Our preliminary results clearly indicate a modification of the calcium activity associated to variations of gravity.
Collapse
Affiliation(s)
- Pierre-Ewen Lecoq
- PMMH, ESPCI Paris - PSL, Paris, 75005, France.
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France.
| | - Chloé Dupuis
- PMMH, ESPCI Paris - PSL, Paris, 75005, France
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France
| | - Xavier Mousset
- PMMH, ESPCI Paris - PSL, Paris, 75005, France
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France
| | | | - Jean-Michel Peyrin
- Neurosciences Paris Seine IBPS, UMR8246, Inserm U1130, Sorbonne University, 4 Place Jussieu, Paris, 75005, France.
| | | |
Collapse
|
21
|
Neikirk K, Stephens DC, Beasley HK, Marshall AG, Gaddy JA, Damo SM, Hinton A. Is space the final frontier for mitochondrial study? Biotechniques 2024; 76:46-51. [PMID: 38084381 DOI: 10.2144/btn-2023-0071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Tweetable abstract This perspective considers several avenues for future research on mitochondrial dynamics, stress, and DNA in outer space.
Collapse
Affiliation(s)
- Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Dominique C Stephens
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Heather K Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven M Damo
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
22
|
Forghani P, Rashid A, Armand LC, Wolfson D, Liu R, Cho HC, Maxwell JT, Jo H, Salaita K, Xu C. Simulated microgravity improves maturation of cardiomyocytes derived from human induced pluripotent stem cells. Sci Rep 2024; 14:2243. [PMID: 38278855 PMCID: PMC10817987 DOI: 10.1038/s41598-024-52453-1] [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: 09/13/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) possess tremendous potential for basic research and translational application. However, these cells structurally and functionally resemble fetal cardiomyocytes, which is a major limitation of these cells. Microgravity can significantly alter cell behavior and function. Here we investigated the effect of simulated microgravity on hiPSC-CM maturation. Following culture under simulated microgravity in a random positioning machine for 7 days, 3D hiPSC-CMs had increased mitochondrial content as detected by a mitochondrial protein and mitochondrial DNA to nuclear DNA ratio. The cells also had increased mitochondrial membrane potential. Consistently, simulated microgravity increased mitochondrial respiration in 3D hiPSC-CMs, as indicated by higher levels of maximal respiration and ATP content, suggesting improved metabolic maturation in simulated microgravity cultures compared with cultures under normal gravity. Cells from simulated microgravity cultures also had improved Ca2+ transient parameters, a functional characteristic of more mature cardiomyocytes. In addition, these cells had improved structural properties associated with more mature cardiomyocytes, including increased sarcomere length, z-disc length, nuclear diameter, and nuclear eccentricity. These findings indicate that microgravity enhances the maturation of hiPSC-CMs at the structural, metabolic, and functional levels.
Collapse
Affiliation(s)
- Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Aysha Rashid
- Biomolecular Chemistry, Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Lawrence C Armand
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - David Wolfson
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Hee Cheol Cho
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Hanjoong Jo
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Khalid Salaita
- Biomolecular Chemistry, Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA.
| |
Collapse
|
23
|
Hwang H, Rampoldi A, Forghani P, Li D, Fite J, Boland G, Maher K, Xu C. Space microgravity increases expression of genes associated with proliferation and differentiation in human cardiac spheres. NPJ Microgravity 2023; 9:88. [PMID: 38071377 PMCID: PMC10710480 DOI: 10.1038/s41526-023-00336-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 11/21/2023] [Indexed: 04/12/2024] Open
Abstract
Efficient generation of cardiomyocytes from human-induced pluripotent stem cells (hiPSCs) is important for their application in basic and translational studies. Space microgravity can significantly change cell activities and function. Previously, we reported upregulation of genes associated with cardiac proliferation in cardiac progenitors derived from hiPSCs that were exposed to space microgravity for 3 days. Here we investigated the effect of long-term exposure of hiPSC-cardiac progenitors to space microgravity on global gene expression. Cryopreserved 3D hiPSC-cardiac progenitors were sent to the International Space Station (ISS) and cultured for 3 weeks under ISS microgravity and ISS 1 G conditions. RNA-sequencing analyses revealed upregulation of genes associated with cardiac differentiation, proliferation, and cardiac structure/function and downregulation of genes associated with extracellular matrix regulation in the ISS microgravity cultures compared with the ISS 1 G cultures. Gene ontology analysis and Kyoto Encyclopedia of Genes and Genomes mapping identified the upregulation of biological processes, molecular function, cellular components, and pathways associated with cell cycle, cardiac differentiation, and cardiac function. Taking together, these results suggest that space microgravity has a beneficial effect on the differentiation and growth of cardiac progenitors.
Collapse
Affiliation(s)
- Hyun Hwang
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | | | - Kevin Maher
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
24
|
Tavakol DN, Nash TR, Kim Y, He S, Fleischer S, Graney PL, Brown JA, Liberman M, Tamargo M, Harken A, Ferrando AA, Amundson S, Garty G, Azizi E, Leong KW, Brenner DJ, Vunjak-Novakovic G. Modeling and countering the effects of cosmic radiation using bioengineered human tissues. Biomaterials 2023; 301:122267. [PMID: 37633022 PMCID: PMC10528250 DOI: 10.1016/j.biomaterials.2023.122267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/28/2023]
Abstract
Cosmic radiation is the most serious risk that will be encountered during the planned missions to the Moon and Mars. There is a compelling need to understand the effects, safety thresholds, and mechanisms of radiation damage in human tissues, in order to develop measures for radiation protection during extended space travel. As animal models fail to recapitulate the molecular changes in astronauts, engineered human tissues and "organs-on-chips" are valuable tools for studying effects of radiation in vitro. We have developed a bioengineered tissue platform for studying radiation damage in individualized settings. To demonstrate its utility, we determined the effects of radiation using engineered models of two human tissues known to be radiosensitive: engineered cardiac tissues (eCT, a target of chronic radiation damage) and engineered bone marrow (eBM, a target of acute radiation damage). We report the effects of high-dose neutrons, a proxy for simulated galactic cosmic rays, on the expression of key genes implicated in tissue responses to ionizing radiation, phenotypic and functional changes in both tissues, and proof-of-principle application of radioprotective agents. We further determined the extent of inflammatory, oxidative stress, and matrix remodeling gene expression changes, and found that these changes were associated with an early hypertrophic phenotype in eCT and myeloid skewing in eBM. We propose that individualized models of human tissues have potential to provide insights into the effects and mechanisms of radiation during deep-space missions and allow testing of radioprotective measures.
Collapse
Affiliation(s)
| | - Trevor R Nash
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Jessie A Brown
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Martin Liberman
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Andrew Harken
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Adolfo A Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Sally Amundson
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Guy Garty
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Elham Azizi
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University, New York, NY 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA; Department of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
25
|
Ren Z, Harriot AD, Mair DB, Chung MK, Lee PHU, Kim DH. Biomanufacturing of 3D Tissue Constructs in Microgravity and their Applications in Human Pathophysiological Studies. Adv Healthc Mater 2023; 12:e2300157. [PMID: 37483106 DOI: 10.1002/adhm.202300157] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/27/2023] [Indexed: 07/25/2023]
Abstract
The growing interest in bioengineering in-vivo-like 3D functional tissues has led to novel approaches to the biomanufacturing process as well as expanded applications for these unique tissue constructs. Microgravity, as seen in spaceflight, is a unique environment that may be beneficial to the tissue-engineering process but cannot be completely replicated on Earth. Additionally, the expense and practical challenges of conducting human and animal research in space make bioengineered microphysiological systems an attractive research model. In this review, published research that exploits real and simulated microgravity to improve the biomanufacturing of a wide range of tissue types as well as those studies that use microphysiological systems, such as organ/tissue chips and multicellular organoids, for modeling human diseases in space are summarized. This review discusses real and simulated microgravity platforms and applications in tissue-engineered microphysiological systems across three topics: 1) application of microgravity to improve the biomanufacturing of tissue constructs, 2) use of tissue constructs fabricated in microgravity as models for human diseases on Earth, and 3) investigating the effects of microgravity on human tissues using biofabricated in vitro models. These current achievements represent important progress in understanding the physiological effects of microgravity and exploiting their advantages for tissue biomanufacturing.
Collapse
Affiliation(s)
- Zhanping Ren
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Peter H U Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, 02720, USA
| | - Deok-Ho Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218, USA
| |
Collapse
|
26
|
Tabury K, Rehnberg E, Baselet B, Baatout S, Moroni L. Bioprinting of Cardiac Tissue in Space: Where Are We? Adv Healthc Mater 2023; 12:e2203338. [PMID: 37312654 PMCID: PMC11469151 DOI: 10.1002/adhm.202203338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/18/2023] [Indexed: 06/15/2023]
Abstract
Bioprinting in space is the next frontier in tissue engineering. In the absence of gravity, novel opportunities arise, as well as new challenges. The cardiovascular system needs particular attention in tissue engineering, not only to develop safe countermeasures for astronauts in future deep and long-term space missions, but also to bring solutions to organ transplantation shortage. In this perspective, the challenges encountered when using bioprinting techniques in space and current gaps that need to be overcome are discussed. The recent developments that have been made in the bioprinting of heart tissues in space and an outlook on potential future bioprinting opportunities in space are described.
Collapse
Affiliation(s)
- Kevin Tabury
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Biomedical EngineeringCollege of Engineering and ComputingUniversity of South CarolinaColumbiaSC29208USA
| | - Emil Rehnberg
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Molecular BiotechnologyGhent UniversityGhent9000Belgium
| | - Bjorn Baselet
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
| | - Sarah Baatout
- Radiology UnitBelgian Nuclear Research CenterBoeretang 200Mol2400Belgium
- Department of Molecular BiotechnologyGhent UniversityGhent9000Belgium
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
27
|
Yau A, Jogdand A, Chen Y. Blood-brain-barrier modeling with tissue chips for research applications in space and on Earth. FRONTIERS IN SPACE TECHNOLOGIES 2023; 4:1176943. [PMID: 38915909 PMCID: PMC11195916 DOI: 10.3389/frspt.2023.1176943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Tissue chip technology has revolutionized biomedical applications and the medical science field for the past few decades. Currently, tissue chips are one of the most powerful research tools aiding in in vitro work to accurately predict the outcome of studies when compared to monolayer two-dimensional (2D) cell cultures. While 2D cell cultures held prominence for a long time, their lack of biomimicry has resulted in a transition to 3D cell cultures, including tissue chips technology, to overcome the discrepancies often seen in in vitro studies. Due to their wide range of applications, different organ systems have been studied over the years, one of which is the blood brain barrier (BBB) which is discussed in this review. The BBB is an incredible protective unit of the body, keeping out pathogens from entering the brain through vasculature. However, there are some microbes and certain diseases that disrupt the function of this barrier which can lead to detrimental outcomes. Over the past few years, various designs of the BBB have been proposed and modeled to study drug delivery and disease modeling on Earth. More recently, researchers have started to utilize tissue chips in space to study the effects of microgravity on human health. BBB tissue chips in space can be a tool to understand function mechanisms and therapeutics. This review addresses the limitations of monolayer cell culture which could be overcome with utilizing tissue chips technology. Current BBB models on Earth and how they are fabricated as well as what influences the BBB cell culture in tissue chips are discussed. Then, this article reviews how application of these technologies together with incorporating biosensors in space would be beneficial to help in predicting a more accurate physiological response in specific tissue or organ chips. Finally, the current platforms used in space and some solutions to overcome some shortcomings for future BBB tissue chip research are also discussed.
Collapse
Affiliation(s)
| | | | - Yupeng Chen
- Nanomedicine Lab, Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
28
|
McKinley S, Taylor A, Peeples C, Jacob M, Khaparde G, Walter Y, Ekpenyong A. Simulated Microgravity-Induced Changes to Drug Response in Cancer Cells Quantified Using Fluorescence Morphometry. Life (Basel) 2023; 13:1683. [PMID: 37629540 PMCID: PMC10455503 DOI: 10.3390/life13081683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Unlike plants that have special gravity-sensing cells, such special cells in animals are yet to be discovered. However, microgravity, the condition of apparent weightlessness, causes bone, muscular and immune system dysfunctions in astronauts following spaceflights. Decades of investigations show correlations between these organ and system-level dysfunctions with changes induced at the cellular level both by simulated microgravity as well as microgravity conditions in outer space. Changes in single bone, muscle and immune cells include morphological abnormalities, altered gene expression, protein expression, metabolic pathways and signaling pathways. These suggest that human cells mount some response to microgravity. However, the implications of such adjustments on many cellular functions and responses are not clear. Here, we addressed the question whether microgravity induces alterations to drug response in cancer cells. We used both adherent cancer cells (T98G) and cancer cells in suspension (K562) to confirm the known effects of simulated microgravity and then treated the K562 cells with common cancer drugs (hydroxyurea and paclitaxel) following 48 h of exposure to simulated microgravity via a NASA-developed rotary cell culture system. Through fluorescence-guided morphometry, we found that microgravity abolished a significant reduction (p < 0.01) in the nuclear-to-cytoplasm ratio of cancer cells treated with hydroxyurea. Our results call for more studies on the impact of microgravity on cellular drug response, in light of the growing need for space medicine, as space exploration grows.
Collapse
Affiliation(s)
- Spencer McKinley
- Biology Department, Creighton University, Omaha, NE 68178, USA; (S.M.); (A.T.); (M.J.); (G.K.)
| | - Adam Taylor
- Biology Department, Creighton University, Omaha, NE 68178, USA; (S.M.); (A.T.); (M.J.); (G.K.)
| | - Conner Peeples
- Physics Department, Creighton University, Omaha, NE 68178, USA; (C.P.); (Y.W.)
| | - Megha Jacob
- Biology Department, Creighton University, Omaha, NE 68178, USA; (S.M.); (A.T.); (M.J.); (G.K.)
| | - Gargee Khaparde
- Biology Department, Creighton University, Omaha, NE 68178, USA; (S.M.); (A.T.); (M.J.); (G.K.)
| | - Yohan Walter
- Physics Department, Creighton University, Omaha, NE 68178, USA; (C.P.); (Y.W.)
| | - Andrew Ekpenyong
- Physics Department, Creighton University, Omaha, NE 68178, USA; (C.P.); (Y.W.)
| |
Collapse
|
29
|
Kuehn BM. Introducing AHA's New President: Joseph C. Wu, MD, PhD, FAHA. J Am Heart Assoc 2023; 12:e031618. [PMID: 37489710 PMCID: PMC10492971 DOI: 10.1161/jaha.123.031618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023]
|
30
|
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: 28] [Impact Index Per Article: 14.0] [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
|
31
|
Elsaesser A, Burr DJ, Mabey P, Urso RG, Billi D, Cockell C, Cottin H, Kish A, Leys N, van Loon JJWA, Mateo-Marti E, Moissl-Eichinger C, Onofri S, Quinn RC, Rabbow E, Rettberg P, de la Torre Noetzel R, Slenzka K, Ricco AJ, de Vera JP, Westall F. Future space experiment platforms for astrobiology and astrochemistry research. NPJ Microgravity 2023; 9:43. [PMID: 37308480 DOI: 10.1038/s41526-023-00292-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Space experiments are a technically challenging but a scientifically important part of astrobiology and astrochemistry research. The International Space Station (ISS) is an excellent example of a highly successful and long-lasting research platform for experiments in space, that has provided a wealth of scientific data over the last two decades. However, future space platforms present new opportunities to conduct experiments with the potential to address key topics in astrobiology and astrochemistry. In this perspective, the European Space Agency (ESA) Topical Team Astrobiology and Astrochemistry (with feedback from the wider scientific community) identifies a number of key topics and summarizes the 2021 "ESA SciSpacE Science Community White Paper" for astrobiology and astrochemistry. We highlight recommendations for the development and implementation of future experiments, discuss types of in situ measurements, experimental parameters, exposure scenarios and orbits, and identify knowledge gaps and how to advance scientific utilization of future space-exposure platforms that are either currently under development or in an advanced planning stage. In addition to the ISS, these platforms include CubeSats and SmallSats, as well as larger platforms such as the Lunar Orbital Gateway. We also provide an outlook for in situ experiments on the Moon and Mars, and welcome new possibilities to support the search for exoplanets and potential biosignatures within and beyond our solar system.
Collapse
Affiliation(s)
- Andreas Elsaesser
- Freie Universitaet Berlin, Department of Physics, Arnimallee 14, 14195, Berlin, Germany.
| | - David J Burr
- Freie Universitaet Berlin, Department of Physics, Arnimallee 14, 14195, Berlin, Germany
| | - Paul Mabey
- Freie Universitaet Berlin, Department of Physics, Arnimallee 14, 14195, Berlin, Germany
| | | | - Daniela Billi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Charles Cockell
- UK Centre for Astrobiology, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Hervé Cottin
- Univ Paris Est Creteil and Université Paris Cité, CNRS, LISA, F-94010, Créteil, France
| | - Adrienne Kish
- Muséum National d'Histoire Naturelle (MNHN), Molécules de Communication et Adaptation des Microorganismes (MCAM), CNRS, 57 rue Cuvier, 75005, Paris, France
| | - Natalie Leys
- Interdisciplinary Biosciences Group, Belgian Nuclear Research Centre, SCK CEN, 2400, Mol, Belgium
| | - 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
| | - Eva Mateo-Marti
- Centro de Astrobiología (CAB), CSIC-INTA, Carretera de Ajalvir km 4, 28850 Torrejón de Ardoz, Madrid, Spain
| | - Christine Moissl-Eichinger
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Silvano Onofri
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Largo dell'Università snc, 01100, Viterbo, Italy
| | | | - Elke Rabbow
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, 51147, Cologne, Germany
| | - Petra Rettberg
- German Aerospace Center (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, 51147, Cologne, Germany
| | - Rosa de la Torre Noetzel
- Instituto Nacional de Técnica Aeroespacial (INTA), Departamento de Observación de la Tierra, 28850 Torrejón de Ardoz, Madrid, Spain
| | - Klaus Slenzka
- KS-3D-3D-Printing and Laser Services, In der Heide 16, 27243, Gross Ippener, Germany
| | | | - Jean-Pierre de Vera
- German Aerospace Center (DLR), Space Operations and Astronaut Training, Microgravity User Support Center (MUSC), Linder Höhe, 51147, Cologne, Germany
| | - Frances Westall
- Centre National de la Recherche Scientifique (CNRS), Centre de Biophysique Moléculaire, Orléans, France
| |
Collapse
|
32
|
Sharma SN, Meller LLT, Sharma AN, Amsterdam EA. Cardiovascular Adaptations of Space Travel: A Systematic Review. Cardiology 2023; 148:434-440. [PMID: 37302388 PMCID: PMC10614241 DOI: 10.1159/000531466] [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: 01/21/2023] [Accepted: 05/31/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Space travel imposes significant gravitational and radiation stress on both cellular and systemic physiology, resulting in myriad cardiovascular changes that have not been fully characterized. METHODS We conducted a systematic review of the cellular and clinical adaptations of the cardiovascular system after exposure to real or simulated space travel in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The PubMed and Cochrane databases were searched in June 2021 for all peer-reviewed articles published since 1950 related to the following search terms entered in separate pairs: "cardiology and space" and "cardiology and astronaut." Only cellular and clinical studies in English concerning the investigation of cardiology and space were included. RESULTS Eighteen studies were identified, comprising 14 clinical and 4 cellular investigations. On the genetic level, pluripotent stem cells in humans and cardiomyocytes in mice displayed increased beat irregularity, with clinical studies revealing a persistent increase in heart rate after space travel. Further cardiovascular adaptations included a higher frequency of orthostatic tachycardia but no evidence of orthostatic hypotension, after return to sea level. Hemoglobin concentration was also consistently decreased after return to Earth. No consistent change in systolic or diastolic blood pressure or any clinically significant arrhythmias were observed during or after space travel. CONCLUSION Changes in oxygen carrying capacity, blood pressure, and post-flight orthostatic tachycardia may serve as reasons to further screen for pre-existing anemic and hypotensive conditions among astronauts.
Collapse
Affiliation(s)
| | - Leo L T Meller
- School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Ajay Nair Sharma
- School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Ezra A Amsterdam
- Division of Cardiology, Department of Internal Medicine, University of California, Davis, California, USA
| |
Collapse
|
33
|
Park JH, Lee JR, Park S, Kim YJ, Yoon JK, Park HS, Hyun J, Joung YK, Lee TI, Bhang SH. Subaqueous 3D stem cell spheroid levitation culture using anti-gravity bioreactor based on sound wave superposition. Biomater Res 2023; 27:51. [PMID: 37208764 DOI: 10.1186/s40824-023-00383-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Recently, various studies have revealed that 3D cell spheroids have several advantages over 2D cells in stem cell culture. However, conventional 3D spheroid culture methods have some disadvantages and limitations such as time required for spheroid formation and complexity of the experimental process. Here, we used acoustic levitation as cell culture platform to overcome the limitation of conventional 3D culture methods. METHODS In our anti-gravity bioreactor, continuous standing sonic waves created pressure field for 3D culture of human mesenchymal stem cells (hMSCs). hMSCs were trapped and aggerated in pressure field and consequently formed spheroids. The structure, viability, gene and protein expression of spheroids formed in the anti-gravity bioreactor were analyzed by electron microscope, immunostaining, polymerase chain reaction, and western blot. We injected hMSC spheroids fabricated by anti-gravity bioreactor into the mouse hindlimb ischemia model. Limb salvage was quantified to evaluate therapeutic efficacy of hMSC spheroids. RESULTS The acoustic levitation in anti-gravity bioreactor made spheroids faster and more compact compared to the conventional hanging drop method, which resulted in the upregulation of angiogenic paracrine factors of hMSCs, such as vascular endothelial growth factor and angiopoietin 2. Injected hMSCs spheroids cultured in the anti-gravity bioreactor exhibited improved therapeutic efficacy, including the degree of limb salvage, capillary formation, and attenuation of fibrosis and inflammation, for mouse hindlimb ischemia model compared to spheroids formed by the conventional hanging drop method. CONCLUSION Our stem cell culture system using acoustic levitation will be proposed as a new platform for the future 3D cell culture system.
Collapse
Affiliation(s)
- Jung Hwan Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ju-Ro Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sungkwon Park
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, 05006, Korea
| | - Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Gyeonggi-Do, Anseong-Si, 17540, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Republic of Korea, Seoul, 02792, Republic of Korea
| | - Tae Il Lee
- Department of Materials Science and Engineering, Gachon University, Gyeonggi-Do, Seongnam-Si, 13120, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
34
|
Theotokis P, Manthou ME, Deftereou TE, Miliaras D, Meditskou S. Addressing Spaceflight Biology through the Lens of a Histologist-Embryologist. Life (Basel) 2023; 13:life13020588. [PMID: 36836946 PMCID: PMC9965490 DOI: 10.3390/life13020588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Embryogenesis and fetal development are highly delicate and error-prone processes in their core physiology, let alone if stress-associated factors and conditions are involved. Space radiation and altered gravity are factors that could radically affect fertility and pregnancy and compromise a physiological organogenesis. Unfortunately, there is a dearth of information examining the effects of cosmic exposures on reproductive and proliferating outcomes with regard to mammalian embryonic development. However, explicit attention has been given to investigations exploring discrete structures and neural networks such as the vestibular system, an entity that is viewed as the sixth sense and organically controls gravity beginning with the prenatal period. The role of the gut microbiome, a newly acknowledged field of research in the space community, is also being challenged to be added in forthcoming experimental protocols. This review discusses the data that have surfaced from simulations or actual space expeditions and addresses developmental adaptations at the histological level induced by an extraterrestrial milieu.
Collapse
Affiliation(s)
- Paschalis Theotokis
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Dimosthenis Miliaras
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Soultana Meditskou
- Laboratory of Histology and Embryology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence:
| |
Collapse
|
35
|
Veliz AL, Mamoun L, Hughes L, Vega R, Holmes B, Monteon A, Bray J, Pecaut MJ, Kearns-Jonker M. Transcriptomic Effects on the Mouse Heart Following 30 Days on the International Space Station. Biomolecules 2023; 13:biom13020371. [PMID: 36830740 PMCID: PMC9953463 DOI: 10.3390/biom13020371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Efforts to understand the impact of spaceflight on the human body stem from growing interest in long-term space travel. Multiple organ systems are affected by microgravity and radiation, including the cardiovascular system. Previous transcriptomic studies have sought to reveal the changes in gene expression after spaceflight. However, little is known about the impact of long-term spaceflight on the mouse heart in vivo. This study focuses on the transcriptomic changes in the hearts of female C57BL/6J mice flown on the International Space Station (ISS) for 30 days. RNA was isolated from the hearts of three flight and three comparable ground control mice and RNA sequencing was performed. Our analyses showed that 1147 transcripts were significantly regulated after spaceflight. The MAPK, PI3K-Akt, and GPCR signaling pathways were predicted to be activated. Transcripts related to cytoskeleton breakdown and organization were upregulated, but no significant change in the expression of extracellular matrix (ECM) components or oxidative stress pathway-associated transcripts occurred. Our results indicate an absence of cellular senescence, and a significant upregulation of transcripts associated with the cell cycle. Transcripts related to cellular maintenance and survival were most affected by spaceflight, suggesting that cardiovascular transcriptome initiates an adaptive response to long-term spaceflight.
Collapse
Affiliation(s)
- Alicia L. Veliz
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lana Mamoun
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lorelei Hughes
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Richard Vega
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Bailey Holmes
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Andrea Monteon
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jillian Bray
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Michael J. Pecaut
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Correspondence:
| |
Collapse
|
36
|
Ma C, Duan X, Lei X. 3D cell culture model: From ground experiment to microgravity study. Front Bioeng Biotechnol 2023; 11:1136583. [PMID: 37034251 PMCID: PMC10080128 DOI: 10.3389/fbioe.2023.1136583] [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: 01/03/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Microgravity has been shown to induce many changes in cell growth and differentiation due to offloading the gravitational strain normally exerted on cells. Although many studies have used two-dimensional (2D) cell culture systems to investigate the effects of microgravity on cell growth, three-dimensional (3D) culture scaffolds can offer more direct indications of the modified cell response to microgravity-related dysregulations compared to 2D culture methods. Thus, knowledge of 3D cell culture is essential for better understanding the in vivo tissue function and physiological response under microgravity conditions. This review discusses the advances in 2D and 3D cell culture studies, particularly emphasizing the role of hydrogels, which can provide cells with a mimic in vivo environment to collect a more natural response. We also summarized recent studies about cell growth and differentiation under real microgravity or simulated microgravity conditions using ground-based equipment. Finally, we anticipate that hydrogel-based 3D culture models will play an essential role in constructing organoids, discovering the causes of microgravity-dependent molecular and cellular changes, improving space tissue regeneration, and developing innovative therapeutic strategies. Future research into the 3D culture in microgravity conditions could lead to valuable therapeutic applications in health and pharmaceuticals.
Collapse
Affiliation(s)
- Chiyuan Ma
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xianglong Duan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
- Second Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, China
- *Correspondence: Xianglong Duan, ; Xiaohua Lei,
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Xianglong Duan, ; Xiaohua Lei,
| |
Collapse
|
37
|
Sy MR, Keefe JA, Sutton JP, Wehrens XHT. Cardiac function, structural, and electrical remodeling by microgravity exposure. Am J Physiol Heart Circ Physiol 2023; 324:H1-H13. [PMID: 36399385 PMCID: PMC9762974 DOI: 10.1152/ajpheart.00611.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Space medicine is key to the human exploration of outer space and pushes the boundaries of science, technology, and medicine. Because of harsh environmental conditions related to microgravity and other factors and hazards in outer space, astronauts and spaceflight participants face unique health and medical challenges, including those related to the heart. In this review, we summarize the literature regarding the effects of spaceflight on cardiac structure and function. We also provide an in-depth review of the literature regarding the effects of microgravity on cardiac calcium handling. Our review can inform future mechanistic and therapeutic studies and is applicable to other physiological states similar to microgravity such as prolonged horizontal bed rest and immobilization.
Collapse
Affiliation(s)
- Mary R Sy
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
| | - Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
| | - Jeffrey P Sutton
- Center for Space Medicine, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas
- Center for Space Medicine, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
38
|
Overbey EG, Das S, Cope H, Madrigal P, Andrusivova Z, Frapard S, Klotz R, Bezdan D, Gupta A, Scott RT, Park J, Chirko D, Galazka JM, Costes SV, Mason CE, Herranz R, Szewczyk NJ, Borg J, Giacomello S. Challenges and considerations for single-cell and spatially resolved transcriptomics sample collection during spaceflight. CELL REPORTS METHODS 2022; 2:100325. [PMID: 36452864 PMCID: PMC9701605 DOI: 10.1016/j.crmeth.2022.100325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) have experienced rapid development in recent years. The findings of spaceflight-based scRNA-seq and SRT investigations are likely to improve our understanding of life in space and our comprehension of gene expression in various cell systems and tissue dynamics. However, compared to their Earth-based counterparts, gene expression experiments conducted in spaceflight have not experienced the same pace of development. Out of the hundreds of spaceflight gene expression datasets available, only a few used scRNA-seq and SRT. In this perspective piece, we explore the growing importance of scRNA-seq and SRT in space biology and discuss the challenges and considerations relevant to robust experimental design to enable growth of these methods in the field.
Collapse
Affiliation(s)
- Eliah G. Overbey
- Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, New York, NY, USA
| | - Saswati Das
- Department of Biochemistry, Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | - Henry Cope
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Pedro Madrigal
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Wellcome Genome Campus, Hinxton, UK
| | - Zaneta Andrusivova
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rebecca Klotz
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Daniela Bezdan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany
- NGS Competence Center Tübingen (NCCT), University of Tübingen, Tübingen, German
- yuri GmbH, Meckenbeuren, Germany
| | | | - Ryan T. Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | | | - Jonathan M. Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Sylvain V. Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Christopher E. Mason
- Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, New York, NY, USA
- The WorldQuant Initiative for Quantitative Prediction, New York, NY, USA
| | - Raul Herranz
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid 28040, Spain
| | - Nathaniel J. Szewczyk
- School of Medicine, University of Nottingham, Derby DE22 3DT, UK
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Joseph Borg
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
39
|
Mu X, He W, Rivera VAM, De Alba RAD, Newman DJ, Zhang YS. Small tissue chips with big opportunities for space medicine. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:150-157. [PMID: 36336360 PMCID: PMC11016463 DOI: 10.1016/j.lssr.2022.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
The spaceflight environment, including microgravity and radiation, may have considerable effects on the health and performance of astronauts, especially for long-duration and Martian missions. Conventional on-ground and in-space experimental approaches have been employed to investigate the comprehensive biological effects of the spaceflight environment. As a class of recently emerging bioengineered in vitro models, tissue chips are characterized by a small footprint, potential automation, and the recapitulation of tissue-level physiology, thus promising to help provide molecular and cellular insights into space medicine. Here, we briefly review the technical advantages of tissue chips and discuss specific on-chip physiological recapitulations. Several tissue chips have been launched into space, and more are poised to come through multi-agency collaborations, implying an increasingly important role of tissue chips in space medicine.
Collapse
Affiliation(s)
- Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, IA 52242, USA
| | - Weishen He
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Victoria Abril Manjarrez Rivera
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Raul Armando Duran De Alba
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Dava J Newman
- MIT Media Lab, Department of Aeronautics and Astronautics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA.
| |
Collapse
|
40
|
Cao X, Weil MM, Wu JC. Clinical Trial in a Dish for Space Radiation Countermeasure Discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:140-149. [PMID: 36336359 PMCID: PMC10947779 DOI: 10.1016/j.lssr.2022.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/30/2022] [Accepted: 05/25/2022] [Indexed: 06/16/2023]
Abstract
NASA aims to return humans to the moon within the next five years and to land humans on Mars in a few decades. Space radiation exposure represents a major challenge to astronauts' health during long-duration missions, as it is linked to increased risks of cancer, cardiovascular dysfunctions, central nervous system (CNS) impairment, and other negative outcomes. Characterization of radiation health effects and developing corresponding countermeasures are high priorities for the preparation of long duration space travel. Due to limitations of animal and cell models, the development of novel physiologically relevant radiation models is needed to better predict these individual risks and bridge gaps between preclinical testing and clinical trials in drug development. "Clinical Trial in a Dish" (CTiD) is now possible with the use of human induced pluripotent stem cells (hiPSCs), offering a powerful tool for drug safety or efficacy testing using patient-specific cell models. Here we review the development and applications of CTiD for space radiation biology and countermeasure studies, focusing on progress made in the past decade.
Collapse
Affiliation(s)
- Xu Cao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA 94305, USA; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael M Weil
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA 94305, USA; Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
41
|
Stem Cell Therapy for Sequestration of Traumatic Brain Injury-Induced Inflammation. Int J Mol Sci 2022; 23:ijms231810286. [PMID: 36142198 PMCID: PMC9499317 DOI: 10.3390/ijms231810286] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of long-term neurological disabilities in the world. TBI is a signature disease for soldiers and veterans, but also affects civilians, including adults and children. Following TBI, the brain resident and immune cells turn into a “reactive” state, characterized by the production of inflammatory mediators that contribute to the development of cognitive deficits. Other injuries to the brain, including radiation exposure, may trigger TBI-like pathology, characterized by inflammation. Currently there are no treatments to prevent or reverse the deleterious consequences of brain trauma. The recognition that TBI predisposes stem cell alterations suggests that stem cell-based therapies stand as a potential treatment for TBI. Here, we discuss the inflamed brain after TBI and radiation injury. We further review the status of stem cells in the inflamed brain and the applications of cell therapy in sequestering inflammation in TBI.
Collapse
|
42
|
Rampoldi A, Forghani P, Li D, Hwang H, Armand LC, Fite J, Boland G, Maxwell J, Maher K, Xu C. Space microgravity improves proliferation of human iPSC-derived cardiomyocytes. Stem Cell Reports 2022; 17:2272-2285. [PMID: 36084640 PMCID: PMC9561632 DOI: 10.1016/j.stemcr.2022.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
In microgravity, cells undergo profound changes in their properties. However, how human cardiac progenitors respond to space microgravity is unknown. In this study, we evaluated the effect of space microgravity on differentiation of human induced pluripotent stem cell (hiPSC)-derived cardiac progenitors compared with 1G cultures on the International Space Station (ISS). Cryopreserved 3D cardiac progenitors were cultured for 3 weeks on the ISS. Compared with 1G cultures, the microgravity cultures had 3-fold larger sphere sizes, 20-fold higher counts of nuclei, and increased expression of proliferation markers. Highly enriched cardiomyocytes generated in space microgravity showed improved Ca2+ handling and increased expression of contraction-associated genes. Short-term exposure (3 days) of cardiac progenitors to space microgravity upregulated genes involved in cell proliferation, survival, cardiac differentiation, and contraction, consistent with improved microgravity cultures at the late stage. These results indicate that space microgravity increased proliferation of hiPSC-cardiomyocytes, which had appropriate structure and function. Cryopreserved 3D hiPSC-cardiac progenitors differentiated efficiently in space Microgravity cultures had increased sphere sizes and cellular proliferation Beating cardiomyocytes in microgravity cultures had improved Ca2+ handling Microgravity cultures had upregulated genes in cardiac contraction
Collapse
Affiliation(s)
- Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Hyun Hwang
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Lawrence Christian Armand
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | | | - Joshua Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kevin Maher
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
43
|
Timilsina S, Kirsch-Mangu T, Werth S, Shepard B, Ma T, Villa-Diaz LG. Enhanced self-renewal of human pluripotent stem cells by simulated microgravity. NPJ Microgravity 2022; 8:22. [PMID: 35787634 PMCID: PMC9253108 DOI: 10.1038/s41526-022-00209-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/22/2022] [Indexed: 11/19/2022] Open
Abstract
A systematic study on the biological effects of simulated microgravity (sµg) on human pluripotent stem cells (hPSC) is still lacking. Here, we used a fast-rotating 2-D clinostat to investigate the sµg effect on proliferation, self-renewal, and cell cycle regulation of hPSCs. We observed significant upregulation of protein translation of pluripotent transcription factors in hPSC cultured in sµg compared to cells cultured in 1g conditions. In addition to a significant increase in expression of telomere elongation genes. Differentiation experiments showed that hPSC cultured in sµg condition were less susceptible to differentiation compared to cells in 1g conditions. These results suggest that sµg enhances hPSC self-renewal. Our study revealed that sµg enhanced the cell proliferation of hPSCs by regulating the expression of cell cycle-associated kinases. RNA-seq analysis indicated that in sµg condition the expression of differentiation and development pathways are downregulated, while multiple components of the ubiquitin proteasome system are upregulated, contributing to an enhanced self-renewal of hPSCs. These effects of sµg were not replicated in human fibroblasts. Taken together, our results highlight pathways and mechanisms in hPSCs vulnerable to microgravity that imposes significant impacts on human health and performance, physiology, and cellular and molecular processes.
Collapse
Affiliation(s)
- S Timilsina
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - T Kirsch-Mangu
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - S Werth
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - B Shepard
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - T Ma
- Department of Computer Science, Engineering, Oakland University, Rochester, MI, 48309, USA
| | - L G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA. .,Department of Bioengineering, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
44
|
Cope H, Willis CR, MacKay MJ, Rutter LA, Toh LS, Williams PM, Herranz R, Borg J, Bezdan D, Giacomello S, Muratani M, Mason CE, Etheridge T, Szewczyk NJ. Routine omics collection is a golden opportunity for European human research in space and analog environments. PATTERNS 2022; 3:100550. [PMID: 36277820 PMCID: PMC9583032 DOI: 10.1016/j.patter.2022.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Iberite F, Gruppioni E, Ricotti L. Skeletal muscle differentiation of human iPSCs meets bioengineering strategies: perspectives and challenges. NPJ Regen Med 2022; 7:23. [PMID: 35393412 PMCID: PMC8991236 DOI: 10.1038/s41536-022-00216-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Although skeletal muscle repairs itself following small injuries, genetic diseases or severe damages may hamper its ability to do so. Induced pluripotent stem cells (iPSCs) can generate myogenic progenitors, but their use in combination with bioengineering strategies to modulate their phenotype has not been sufficiently investigated. This review highlights the potential of this combination aimed at pushing the boundaries of skeletal muscle tissue engineering. First, the overall organization and the key steps in the myogenic process occurring in vivo are described. Second, transgenic and non-transgenic approaches for the myogenic induction of human iPSCs are compared. Third, technologies to provide cells with biophysical stimuli, biomaterial cues, and biofabrication strategies are discussed in terms of recreating a biomimetic environment and thus helping to engineer a myogenic phenotype. The embryonic development process and the pro-myogenic role of the muscle-resident cell populations in co-cultures are also described, highlighting the possible clinical applications of iPSCs in the skeletal muscle tissue engineering field.
Collapse
Affiliation(s)
- Federica Iberite
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy. .,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.
| | - Emanuele Gruppioni
- Centro Protesi INAIL, Istituto Nazionale per l'Assicurazione contro gli Infortuni sul Lavoro, 40054, Vigorso di Budrio (BO), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy
| |
Collapse
|
46
|
Abstract
The direct (eg, radiation, microgravity) and indirect (eg, lifestyle perturbations) effects of spaceflight extend across multiple systems resulting in whole-organism cardiovascular deconditioning. For over 50 years, National Aeronautics and Space Administration has continually enhanced a countermeasures program designed to characterize and offset the adverse cardiovascular consequences of spaceflight. In this review, we provide a historical overview of research evaluating the effects of spaceflight on cardiovascular health in astronauts and outline mechanisms underpinning spaceflight-related cardiovascular alterations. We also discuss how spaceflight could be leveraged for aging, industry, and model systems such as human induced pluripotent stem cell-derived cardiomyocytes, organoid, and organ-on-a-chip technologies. Finally, we outline the increasing opportunities for scientists and clinicians to engage in cardiovascular research in space and on Earth.
Collapse
Affiliation(s)
- Jessica M Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY (J.M.S.).,Weill Cornell Medical College, New York, NY (J.M.S.)
| | | | - Lianne Dolan
- Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada (L.D.)
| | | |
Collapse
|
47
|
Jirak P, Mirna M, Rezar R, Motloch LJ, Lichtenauer M, Jordan J, Binneboessel S, Tank J, Limper U, Jung C. How spaceflight challenges human cardiovascular health. Eur J Prev Cardiol 2022; 29:1399-1411. [PMID: 35148376 DOI: 10.1093/eurjpc/zwac029] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/08/2022] [Accepted: 02/06/2022] [Indexed: 11/14/2022]
Abstract
The harsh environmental conditions in space, particularly weightlessness and radiation exposure, can negatively affect cardiovascular function and structure. In the future, preventive cardiology will be crucial in enabling safe space travel. Indeed, future space missions destined to the Moon and from there to Mars will create new challenges to cardiovascular health while limiting medical management. Moreover, commercial spaceflight evolves rapidly such that older persons with cardiovascular risk factors will be exposed to space conditions. This review provides an overview on studies conducted in space and in terrestrial models, particularly head-down bedrest studies. These studies showed that weightlessness elicits a fluid shift towards the head, which likely predisposes to the spaceflight-associated neuro-ocular syndrome, neck vein thrombosis, and orthostatic intolerance after return to Earth. Moreover, cardiovascular unloading produces cardiopulmonary deconditioning which may be associated with cardiac atrophy. In addition to limiting physical performance, the mechanism further worsens orthostatic tolerance after return to Earth. Finally, space conditions may directly affect vascular health, however, the clinical relevance of these findings in terms of morbidity and mortality is unknown. Targeted preventive measures, which are referred to as countermeasures in aerospace medicine, and technologies to identify vascular risks early on will be required to maintain cardiovascular performance and health during future space missions.
Collapse
Affiliation(s)
- Peter Jirak
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Moritz Mirna
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Richard Rezar
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas J Motloch
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Michael Lichtenauer
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.,Medical Faculty, University of Cologne, Germany
| | - Stephan Binneboessel
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Germany
| | - Jens Tank
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Ulrich Limper
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.,Department of Anaesthesiology and Critical Care Medicine, Merheim Medical Center, Witten/Herdecke University, Cologne, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Germany
| |
Collapse
|
48
|
Criswell T, Swart C, Stoudemire J, Brockbank K, Floren M, Eaker S, Hunsberger J. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:107-113. [PMID: 36239619 PMCID: PMC9562819 DOI: 10.1093/stcltm/szab025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/07/2021] [Indexed: 11/20/2022] Open
Abstract
Advances in regenerative medicine manufacturing continue to be a priority for achieving the full commercial potential of important breakthrough therapies. Equally important will be the establishment of distribution chains that support the transport of live cells and engineered tissues and organs resulting from these advanced biomanufacturing processes. The importance of a well-managed distribution chain for products requiring specialized handling procedures was highlighted during the COVID-19 pandemic and serves as a reminder of the critical role of logistics and distribution in the success of breakthrough therapies. This perspective article will provide insight into current practices and future considerations for creating global distribution chains that facilitate the successful deployment of regenerative medicine therapies to the vast number of patients that would benefit from them worldwide.
Collapse
Affiliation(s)
- Tracy Criswell
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Corresponding author: Tracy Criswell, PhD, Wake Forest School of Medicine, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC 27101, USA. Tel: 336-713-1615;
| | | | | | | | | | | | | |
Collapse
|
49
|
Sharma A, Clemens RA, Garcia O, Taylor DL, Wagner NL, Shepard KA, Gupta A, Malany S, Grodzinsky AJ, Kearns-Jonker M, Mair DB, Kim DH, Roberts MS, Loring JF, Hu J, Warren LE, Eenmaa S, Bozada J, Paljug E, Roth M, Taylor DP, Rodrigue G, Cantini P, Smith AW, Giulianotti MA, Wagner WR. Biomanufacturing in low Earth orbit for regenerative medicine. Stem Cell Reports 2021; 17:1-13. [PMID: 34971562 PMCID: PMC8758939 DOI: 10.1016/j.stemcr.2021.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Research in low Earth orbit (LEO) has become more accessible. The 2020 Biomanufacturing in Space Symposium reviewed space-based regenerative medicine research and discussed leveraging LEO to advance biomanufacturing for regenerative medicine applications. The symposium identified areas where financial investments could stimulate advancements overcoming technical barriers. Opportunities in disease modeling, stem-cell-derived products, and biofabrication were highlighted. The symposium will initiate a roadmap to a sustainable market for regenerative medicine biomanufacturing in space. This perspective summarizes the 2020 Biomanufacturing in Space Symposium, highlights key biomanufacturing opportunities in LEO, and lays the framework for a roadmap to regenerative medicine biomanufacturing in space.
Collapse
Affiliation(s)
- Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | | | - Orquidea Garcia
- Johnson & Johnson 3D Printing Innovation & Customer Solutions, Johnson & Johnson Services, Inc., Irvine, CA, USA
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Kelly A Shepard
- California Institute for Regenerative Medicine, Oakland, CA, USA
| | | | - Siobhan Malany
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Alan J Grodzinsky
- Departments of Biological Engineering, Mechanical Engineering and Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mary Kearns-Jonker
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael S Roberts
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA
| | | | - Jianying Hu
- Center for Computational Health IBM Research, Yorktown Heights, New York, NY, USA
| | - Lara E Warren
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA
| | - Sven Eenmaa
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA
| | - Joe Bozada
- Joseph M. Katz Graduate School of Business, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Paljug
- Joseph M. Katz Graduate School of Business, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Gary Rodrigue
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA
| | - Patrick Cantini
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Amelia W Smith
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA
| | - Marc A Giulianotti
- Center for the Advancement of Science in Space, Inc, Melbourne, FL, USA.
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Departments of Surgery, Bioengineering, Chemical Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
Basirun C, Ferlazzo ML, Howell NR, Liu GJ, Middleton RJ, Martinac B, Narayanan SA, Poole K, Gentile C, Chou J. Microgravity × Radiation: A Space Mechanobiology Approach Toward Cardiovascular Function and Disease. Front Cell Dev Biol 2021; 9:750775. [PMID: 34778261 PMCID: PMC8586646 DOI: 10.3389/fcell.2021.750775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, there has been an increasing interest in space exploration, supported by the accelerated technological advancements in the field. This has led to a new potential environment that humans could be exposed to in the very near future, and therefore an increasing request to evaluate the impact this may have on our body, including health risks associated with this endeavor. A critical component in regulating the human pathophysiology is represented by the cardiovascular system, which may be heavily affected in these extreme environments of microgravity and radiation. This mini review aims to identify the impact of microgravity and radiation on the cardiovascular system. Being able to understand the effect that comes with deep space explorations, including that of microgravity and space radiation, may also allow us to get a deeper understanding of the heart and ultimately our own basic physiological processes. This information may unlock new factors to consider with space exploration whilst simultaneously increasing our knowledge of the cardiovascular system and potentially associated diseases.
Collapse
Affiliation(s)
- Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Melanie L. Ferlazzo
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
- Inserm, U1296 Unit, Radiation: Defense, Health and Environment, Centre Léon Bérard, Lyon, France
| | - Nicholas R. Howell
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
- Discipline of Medical Imaging and Radiation Sciences, Faculty of Medicine and Health, Brain and Mind Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Ryan J. Middleton
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - S. Anand Narayanan
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, United States
| | - Kate Poole
- EMBL Australia Node in Single Molecule Science, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|