51
|
Prasad B, Grimm D, Strauch SM, Erzinger GS, Corydon TJ, Lebert M, Magnusson NE, Infanger M, Richter P, Krüger M. Influence of Microgravity on Apoptosis in Cells, Tissues, and Other Systems In Vivo and In Vitro. Int J Mol Sci 2020; 21:E9373. [PMID: 33317046 PMCID: PMC7764784 DOI: 10.3390/ijms21249373] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
All life forms have evolved under the constant force of gravity on Earth and developed ways to counterbalance acceleration load. In space, shear forces, buoyance-driven convection, and hydrostatic pressure are nullified or strongly reduced. When subjected to microgravity in space, the equilibrium between cell architecture and the external force is disturbed, resulting in changes at the cellular and sub-cellular levels (e.g., cytoskeleton, signal transduction, membrane permeability, etc.). Cosmic radiation also poses great health risks to astronauts because it has high linear energy transfer values that evoke complex DNA and other cellular damage. Space environmental conditions have been shown to influence apoptosis in various cell types. Apoptosis has important functions in morphogenesis, organ development, and wound healing. This review provides an overview of microgravity research platforms and apoptosis. The sections summarize the current knowledge of the impact of microgravity and cosmic radiation on cells with respect to apoptosis. Apoptosis-related microgravity experiments conducted with different mammalian model systems are presented. Recent findings in cells of the immune system, cardiovascular system, brain, eyes, cartilage, bone, gastrointestinal tract, liver, and pancreas, as well as cancer cells investigated under real and simulated microgravity conditions, are discussed. This comprehensive review indicates the potential of the space environment in biomedical research.
Collapse
Affiliation(s)
- Binod Prasad
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; (D.G.); (T.J.C.)
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Sebastian M. Strauch
- Postgraduate Program in Health and Environment, University of Joinville Region, Rua Paulo Malschitzki, 10 - Zona Industrial Norte, Joinville, SC 89219-710, Brazil; (S.M.S.); (G.S.E.)
| | - Gilmar Sidnei Erzinger
- Postgraduate Program in Health and Environment, University of Joinville Region, Rua Paulo Malschitzki, 10 - Zona Industrial Norte, Joinville, SC 89219-710, Brazil; (S.M.S.); (G.S.E.)
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; (D.G.); (T.J.C.)
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus N, Denmark
| | - Michael Lebert
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
- Space Biology Unlimited SAS, 24 Cours de l’Intendance, 33000 Bordeaux, France
| | - Nils E. Magnusson
- Diabetes and Hormone Diseases, Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark;
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Peter Richter
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| |
Collapse
|
52
|
Paul AM, Mhatre SD, Cekanaviciute E, Schreurs AS, Tahimic CGT, Globus RK, Anand S, Crucian BE, Bhattacharya S. Neutrophil-to-Lymphocyte Ratio: A Biomarker to Monitor the Immune Status of Astronauts. Front Immunol 2020; 11:564950. [PMID: 33224136 PMCID: PMC7667275 DOI: 10.3389/fimmu.2020.564950] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
A comprehensive understanding of spaceflight factors involved in immune dysfunction and the evaluation of biomarkers to assess in-flight astronaut health are essential goals for NASA. An elevated neutrophil-to-lymphocyte ratio (NLR) is a potential biomarker candidate, as leukocyte differentials are altered during spaceflight. In the reduced gravity environment of space, rodents and astronauts displayed elevated NLR and granulocyte-to-lymphocyte ratios (GLR), respectively. To simulate microgravity using two well-established ground-based models, we cultured human whole blood-leukocytes in high-aspect rotating wall vessels (HARV-RWV) and used hindlimb unloaded (HU) mice. Both HARV-RWV simulation of leukocytes and HU-exposed mice showed elevated NLR profiles comparable to spaceflight exposed samples. To assess mechanisms involved, we found the simulated microgravity HARV-RWV model resulted in an imbalance of redox processes and activation of myeloperoxidase-producing inflammatory neutrophils, while antioxidant treatment reversed these effects. In the simulated microgravity HU model, mitochondrial catalase-transgenic mice that have reduced oxidative stress responses showed reduced neutrophil counts, NLR, and a dampened release of selective inflammatory cytokines compared to wildtype HU mice, suggesting simulated microgravity induced oxidative stress responses that triggered inflammation. In brief, both spaceflight and simulated microgravity models caused elevated NLR, indicating this as a potential biomarker for future in-flight immune health monitoring.
Collapse
Affiliation(s)
- Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Universities Space Research Association, Columbia, MD, United States
| | - Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,COSMIAC Research Center, University of New Mexico, Albuquerque, NM, United States.,KBR, Houston, TX, United States
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,COSMIAC Research Center, University of New Mexico, Albuquerque, NM, United States.,Department of Biology, University of North Florida, Jacksonville, FL, United States
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| | - Sulekha Anand
- Department of Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Brian E Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Science Center, Houston, TX, United States
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| |
Collapse
|
53
|
Musci RV, Walsh MA, Konopka AR, Wolff CA, Peelor FF, Reiser RF, Santangelo KS, Hamilton KL. The Dunkin Hartley Guinea Pig Is a Model of Primary Osteoarthritis That Also Exhibits Early Onset Myofiber Remodeling That Resembles Human Musculoskeletal Aging. Front Physiol 2020; 11:571372. [PMID: 33192568 PMCID: PMC7658338 DOI: 10.3389/fphys.2020.571372] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle dysfunction, articular cartilage degeneration, and bone loss occur essentially in parallel during aging. Mechanisms contributing to this systemic musculoskeletal decline remain incompletely understood, limiting progress toward developing effective therapeutics. Because the progression of human musculoskeletal aging is slow, researchers rely on rodent models to identify mechanisms and test interventions. The Dunkin Hartley guinea pig is an outbred strain that begins developing primary osteoarthritis by 4 months of age with a progression and pathology similar to aging humans. The purpose of this study was to determine if skeletal muscle remodeling during the progression of osteoarthritis in these guinea pigs resembles musculoskeletal aging in humans. We compared Dunkin Hartley guinea pigs to Strain 13 guinea pigs, which develop osteoarthritis much later in the lifespan. We measured myofiber type and size, muscle density, and long-term fractional protein synthesis rates of the gastrocnemius and soleus muscles in 5, 9, and 15-month-old guinea pigs. There was an age-related decline in skeletal muscle density, a greater proportion of smaller myofibers, and a decline in type II concomitant with a rise in type I myofibers in the gastrocnemius muscles from Dunkin Hartley guinea pigs only. These changes were accompanied by age-related declines in myofibrillar and mitochondrial protein synthesis in the gastrocnemius and soleus. Collectively, these findings suggest Dunkin Hartley guinea pigs experience myofiber remodeling alongside the progression of osteoarthritis, consistent with human musculoskeletal aging. Thus, Dunkin Hartley guinea pigs may be a model to advance discovery and therapeutic development for human musculoskeletal aging.
Collapse
Affiliation(s)
- Robert V Musci
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
| | - Maureen A Walsh
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
| | - Adam R Konopka
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,GRECC, William S Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Christopher A Wolff
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Raoul F Reiser
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Karyn L Hamilton
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States.,Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
54
|
Approaching Gravity as a Continuum Using the Rat Partial Weight-Bearing Model. Life (Basel) 2020; 10:life10100235. [PMID: 33049988 PMCID: PMC7599661 DOI: 10.3390/life10100235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/30/2020] [Accepted: 10/03/2020] [Indexed: 12/12/2022] Open
Abstract
For decades, scientists have relied on animals to understand the risks and consequences of space travel. Animals remain key to study the physiological alterations during spaceflight and provide crucial information about microgravity-induced changes. While spaceflights may appear common, they remain costly and, coupled with limited cargo areas, do not allow for large sample sizes onboard. In 1979, a model of hindlimb unloading (HU) was successfully created to mimic microgravity and has been used extensively since its creation. Four decades later, the first model of mouse partial weight-bearing (PWB) was developed, aiming at mimicking partial gravity environments. Return to the Lunar surface for astronauts is now imminent and prompted the need for an animal model closer to human physiology; hence in 2018, our laboratory created a new model of PWB for adult rats. In this review, we will focus on the rat model of PWB, from its conception to the current state of knowledge. Additionally, we will address how this new model, used in conjunction with HU, will help implement new paradigms allowing scientists to anticipate the physiological alterations and needs of astronauts. Finally, we will discuss the outstanding questions and future perspectives in space research and propose potential solutions using the rat PWB model.
Collapse
|
55
|
Zhu X, Yao T, Wang R, Guo S, Wang X, Zhou Z, Zhang Y, Zhuo X, Wang R, Li JZ, Liu T, Kong X. IRF4 in Skeletal Muscle Regulates Exercise Capacity via PTG/Glycogen Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001502. [PMID: 33042761 PMCID: PMC7539189 DOI: 10.1002/advs.202001502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/09/2020] [Indexed: 06/11/2023]
Abstract
Exercise-induced fatigue and exhaustion are interesting areas for many researchers. Muscle glycogen is critical for physical performance. However, how glycogen metabolism is manipulated during exercise is not very clear. The aim here is to assess the impact of interferon regulatory factor 4 (IRF4) on skeletal muscle glycogen and subsequent regulation of exercise capacity. Skeletal muscle-specific IRF4 knockout mice show normal body weight and insulin sensitivity, but better exercise capacity and increased glycogen content with unaltered triglyceride levels compared to control mice on chow diet. In contrast, mice overexpression of IRF4 displays decreased exercise capacity and lower glycogen content. Mechanistically, IRF4 regulates glycogen-associated regulatory subunit protein targeting to glycogen (PTG) to manipulate glucose metabolism in skeletal muscle. Knockdown of PTG can reverse the effects imposed by the absence of IRF4 in vivo. These studies reveal a regulatory pathway including IRF4/PTG/glycogen synthesis on controlling exercise capacity.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Division of Pediatric EndocrinologyDepartment of PediatricsUCLA Children's Discovery and Innovation InstituteDavid Geffen School of Medicine at UCLALos AngelesCA90095USA
- Department of Endocrinology and MetabolismZhongshan HospitalFudan UniversityShanghai200032P. R. China
- Fudan Institute for Metabolic DiseaseFudan UniversityShanghai200032P. R. China
| | - Ting Yao
- Division of Pediatric EndocrinologyDepartment of PediatricsUCLA Children's Discovery and Innovation InstituteDavid Geffen School of Medicine at UCLALos AngelesCA90095USA
| | - Ru Wang
- School of KinesiologyKey Laboratory of Exercise and Health Sciences of Ministry of EducationShanghai University of SportShanghai200438P. R. China
| | - Shanshan Guo
- School of KinesiologyKey Laboratory of Exercise and Health Sciences of Ministry of EducationShanghai University of SportShanghai200438P. R. China
| | - Xin Wang
- Division of Pediatric EndocrinologyDepartment of PediatricsUCLA Children's Discovery and Innovation InstituteDavid Geffen School of Medicine at UCLALos AngelesCA90095USA
- Department of Internal MedicineHarbin Medical University Cancer HospitalHarbin Medical UniversityNo. 150 Haping ST, Nangang DistrictHarbinHeilongjiang150081P. R. China
| | - Zhenqi Zhou
- Department of MedicineDivision of Endocrinology, Diabetes and HypertensionDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCA90095USA
| | - Yan Zhang
- School of Life SciencesFudan UniversityShanghai200032P. R. China
| | - Xiaozhen Zhuo
- Department of CardiologyThe First Affiliated HospitalXi'an Jiaotong UniversityXi'anShanxi710061P. R. China
| | - Ruitao Wang
- Department of Internal MedicineHarbin Medical University Cancer HospitalHarbin Medical UniversityNo. 150 Haping ST, Nangang DistrictHarbinHeilongjiang150081P. R. China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic DiseaseDepartment of Biochemistry and Molecular BiologyThe Key Laboratory of Human Functional Genomics of Jiangsu ProvinceNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Tiemin Liu
- State Key Laboratory of Genetic EngineeringDepartment of Endocrinology and Metabolism and School of Life SciencesZhongshan HospitalFudan UniversityShanghai200032P. R. China
- Institute of Metabolism and Integrative Biologyand Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghai200032P. R. China
- Human Phenome InstituteFudan UniversityShanghai200032P. R. China
| | - Xingxing Kong
- Division of Pediatric EndocrinologyDepartment of PediatricsUCLA Children's Discovery and Innovation InstituteDavid Geffen School of Medicine at UCLALos AngelesCA90095USA
| |
Collapse
|
56
|
Circulating Ouabain Modulates Expression of Claudins in Rat Intestine and Cerebral Blood Vessels. Int J Mol Sci 2020; 21:ijms21145067. [PMID: 32709081 PMCID: PMC7404321 DOI: 10.3390/ijms21145067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/20/2022] Open
Abstract
The ability of exogenous low ouabain concentrations to affect claudin expression and therefore epithelial barrier properties was demonstrated previously in cultured cell studies. We hypothesized that chronic elevation of circulating ouabain in vivo can affect the expression of claudins and tight junction permeability in different tissues. We tested this hypothesis in rats intraperitoneally injected with ouabain (1 μg/kg) for 4 days. Rat jejunum, colon and brain frontal lobes, which are variable in the expressed claudins and tight junction permeability, were examined. Moreover, the porcine jejunum cell line IPEC-J2 was studied. In IPEC-J2-cells, ouabain (10 nM, 19 days of incubation) stimulated epithelial barrier formation, increased transepithelial resistance and the level of cSrc-kinase activation by phosphorylation, accompanied with an increased expression of claudin-1, -5 and down-regulation of claudin-12; the expression of claudin-3, -4, -8 and tricellulin was not changed. In the jejunum, chronic ouabain increased the expression of claudin-1, -3 and -5 without an effect on claudin-2 and -4 expression. In the colon, only down-regulation of claudin-3 was observed. Chronic ouabain protected the intestine transepithelial resistance against functional injury induced by lipopolysaccharide treatment or by modeled acute microgravity; this regulation was most pronounced in the jejunum. Claudin-1 was also up-regulated in cerebral blood vessels. This was associated with reduction of claudin-3 expression while the expression of claudin-5 and occludin was not affected. Altogether, our results confirm that circulating ouabain can functionally and tissue-specifically affect barrier properties of epithelial and endothelial tissues via Na,K-ATPase-mediated modulation of claudins expression.
Collapse
|
57
|
Differences in the Role of HDACs 4 and 5 in the Modulation of Processes Regulating MAFbx and MuRF1 Expression during Muscle Unloading. Int J Mol Sci 2020; 21:ijms21134815. [PMID: 32646070 PMCID: PMC7370043 DOI: 10.3390/ijms21134815] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 02/06/2023] Open
Abstract
Unloading leads to skeletal muscle atrophy via the upregulation of MuRF-1 and MAFbx E3-ligases expression. Reportedly, histone deacetylases (HDACs) 4 and 5 may regulate the expression of MuRF1 and MAFbx. To examine the HDAC-dependent mechanisms involved in the control of E3-ubiquitin ligases expression at the early stages of muscle unloading we used HDACs 4 and 5 inhibitor LMK-235 and HDAC 4 inhibitor Tasqinimod (Tq). Male Wistar rats were divided into four groups (eight rats per group): nontreated control (C), three days of unloading/hindlimb suspension (HS) and three days HS with HDACs inhibitor LMK-235 (HSLMK) or Tq (HSTq). Treatment with LMK-235 diminished unloading-induced of MAFbx, myogenin (MYOG), ubiquitin and calpain-1 mRNA expression (p < 0.05). Tq administration had no effect on the expression of E3-ligases. The mRNA expression of MuRF1 and MAFbx was significantly increased in both HS and HSTq groups (1.5 and 4.0 folds, respectively; p < 0.05) when compared with the C group. It is concluded that during three days of muscle unloading: (1) the HDACs 4 and 5 participate in the regulation of MAFbx expression as well as the expression of MYOG, ubiquitin and calpain-1; (2) the inhibition of HDAC 4 has no effect on MAFbx expression. Therefore, HDAC 5 is perhaps more important for the regulation of MAFbx expression than HDAC 4.
Collapse
|
58
|
Kermorgant M, Nasr N, Czosnyka M, Arvanitis DN, Hélissen O, Senard JM, Pavy-Le Traon A. Impacts of Microgravity Analogs to Spaceflight on Cerebral Autoregulation. Front Physiol 2020; 11:778. [PMID: 32719617 PMCID: PMC7350784 DOI: 10.3389/fphys.2020.00778] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
It is well known that exposure to microgravity in astronauts leads to a plethora physiological responses such as headward fluid shift, body unloading, and cardiovascular deconditioning. When astronauts return to Earth, some encounter problems related to orthostatic intolerance. An impaired cerebral autoregulation (CA), which could be compromised by the effects of microgravity, has been proposed as one of the mechanisms responsible for orthostatic intolerance. CA is a homeostatic mechanism that maintains cerebral blood flow for any variations in cerebral perfusion pressure by adapting the vascular tone and cerebral vessel diameter. The ground-based models of microgravity are useful tools for determining the gravitational impact of spaceflight on human body. The head-down tilt bed rest (HDTBR), where the subject remains in supine position at -6 degrees for periods ranging from few days to several weeks is the most commonly used ground-based model of microgravity for cardiovascular deconditioning. head-down bed rest (HDBR) is able to replicate cephalic fluid shift, immobilization, confinement, and inactivity. Dry immersion (DI) model is another approach where the subject remains immersed in thermoneutral water covered with an elastic waterproof fabric separating the subject from the water. Regarding DI, this analog imitates absence of any supporting structure for the body, centralization of body fluids, immobilization and hypokinesia observed during spaceflight. However, little is known about the impact of microgravity on CA. Here, we review the fundamental principles and the different mechanisms involved in CA. We also consider the different approaches in order to assess CA. Finally, we focus on the effects of short- and long-term spaceflight on CA and compare these findings with two specific analogs to microgravity: HDBR and DI.
Collapse
Affiliation(s)
- Marc Kermorgant
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
| | - Nathalie Nasr
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
- Department of Neurology, Institute for Neurosciences, Toulouse University Hospital, Toulouse, France
| | - Marek Czosnyka
- Brain Physics Laboratory, Division of Neurosurgery, Department of Clinical Neurosciences, Cambridge University Hospital, Cambridge, United Kingdom
- Institute of Electronic Systems, Warsaw University of Technology, Warsaw, Poland
| | - Dina N. Arvanitis
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
| | - Ophélie Hélissen
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
| | - Jean-Michel Senard
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
- Department of Clinical Pharmacology, Toulouse University Hospital, Toulouse, France
| | - Anne Pavy-Le Traon
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), Toulouse, France
- Department of Neurology, Institute for Neurosciences, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
59
|
Skeletal Muscle Na,K-ATPase as a Target for Circulating Ouabain. Int J Mol Sci 2020; 21:ijms21082875. [PMID: 32326025 PMCID: PMC7215781 DOI: 10.3390/ijms21082875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 02/07/2023] Open
Abstract
While the role of circulating ouabain-like compounds in the cardiovascular and central nervous systems, kidney and other tissues in health and disease is well documented, little is known about its effects in skeletal muscle. In this study, rats were intraperitoneally injected with ouabain (0.1-10 µg/kg for 4 days) alone or with subsequent injections of lipopolysaccharide (1 mg/kg). Some rats were also subjected to disuse for 6 h by hindlimb suspension. In the diaphragm muscle, chronic ouabain (1 µg/kg) hyperpolarized resting potential of extrajunctional membrane due to specific increase in electrogenic transport activity of the 2 Na,K-ATPase isozyme and without changes in 1 and 2 Na,K-ATPase protein content. Ouabain (10-20 nM), acutely applied to isolated intact diaphragm muscle from not injected rats, hyperpolarized the membrane to a similar extent. Chronic ouabain administration prevented lipopolysaccharide-induced (diaphragm muscle) or disuse-induced (soleus muscle) depolarization of the extrajunctional membrane. No stimulation of the 1 Na,K-ATPase activity in human red blood cells, purified lamb kidney and Torpedo membrane preparations by low ouabain concentrations was observed. Our results suggest that skeletal muscle electrogenesis is subjected to regulation by circulating ouabain via the 2 Na,K-ATPase isozyme that could be important for adaptation of this tissue to functional impairment.
Collapse
|
60
|
Qaisar R, Karim A, Elmoselhi AB. Muscle unloading: A comparison between spaceflight and ground-based models. Acta Physiol (Oxf) 2020; 228:e13431. [PMID: 31840423 DOI: 10.1111/apha.13431] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Prolonged unloading of skeletal muscle, a common outcome of events such as spaceflight, bed rest and hindlimb unloading, can result in extensive metabolic, structural and functional changes in muscle fibres. With advancement in investigations of cellular and molecular mechanisms, understanding of disuse muscle atrophy has significantly increased. However, substantial gaps exist in our understanding of the processes dictating muscle plasticity during unloading, which prevent us from developing effective interventions to combat muscle loss. This review aims to update the status of knowledge and underlying mechanisms leading to cellular and molecular changes in skeletal muscle during unloading. We have also discussed advances in the understanding of contractile dysfunction during spaceflights and in ground-based models of muscle unloading. Additionally, we have elaborated on potential therapeutic interventions that show promising results in boosting muscle mass and strength during mechanical unloading. Finally, we have identified key gaps in our knowledge as well as possible research direction for the future.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Asima Karim
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Adel B. Elmoselhi
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
- Department of Physiology Michigan State University East Lansing MI USA
| |
Collapse
|
61
|
Kravtsova VV, Vilchinskaya NA, Rozlomii VL, Shenkman BS, Krivoi II. Low Ouabain Doses and AMP-Activated Protein Kinase as Factors Supporting Electrogenesis in Skeletal Muscle. BIOCHEMISTRY (MOSCOW) 2019; 84:1085-1092. [PMID: 31693468 DOI: 10.1134/s0006297919090116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many motor disorders are associated with depolarization of the membrane of skeletal muscle fibers due to the impaired functioning of Na,K-ATPase. Here, we studied the role of ouabain (specific Na,K-ATPase ligand) and AMP-activated protein kinase (key regulator of muscle metabolism) in the maintenance of muscle electrogenesis; the levels of these endogenous factors are directly related to the motor activity. After 4-day intraperitoneal administration of ouabain (1 µg/kg daily), a hyperpolarization of sarcolemma was registered in isolated rat diaphragm muscles due to an increase in the electrogenic activity of Na,K-ATPase. In acute experiments, addition of nanomolar ouabain concentrations to the bathing solution resulted in the muscle membrane hyperpolarization within 15 min. The effect of ouabain reversed to membrane depolarization with the increase in the external potassium concentration. It is possible that Na,K-ATPase activation by ouabain may be regulated by such factors as specific subcellular location, interaction with molecular partners, and changes in the ionic balance. Preventive administration of the AMP-activated protein kinase activator AICAR (5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside; 400 mg/kg body weight daily for 7 days) in chronic experiments resulted in the stabilization of the endplate structure and abolishment of depolarization of the rat soleus muscle membrane caused by the motor activity cessation. The obtained data can be useful for creating approaches for correction of muscle dysfunction, especially at the early stages, prior to the development of muscle atrophy.
Collapse
Affiliation(s)
- V V Kravtsova
- St. Petersburg State University, Department of General Physiology, St. Petersburg, 199034, Russia
| | - N A Vilchinskaya
- Institute of Biomedical Problems, Laboratory of Myology, Russian Academy of Sciences, Moscow, 123007, Russia
| | - V L Rozlomii
- St. Petersburg State University, Department of General Physiology, St. Petersburg, 199034, Russia
| | - B S Shenkman
- Institute of Biomedical Problems, Laboratory of Myology, Russian Academy of Sciences, Moscow, 123007, Russia
| | - I I Krivoi
- St. Petersburg State University, Department of General Physiology, St. Petersburg, 199034, Russia.
| |
Collapse
|
62
|
Wu Y, Yue Z, Wang Q, Lv Q, Liu H, Bai Y, Li S, Xie M, Bao J, Ma J, Zhu X, Wang Z. BK Ca compensates impaired coronary vasoreactivity through RhoA/ROCK pathway in hind-limb unweighted rats. FASEB J 2019; 33:13358-13366. [PMID: 31530101 DOI: 10.1096/fj.201901273r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Previous studies have demonstrated cardiac and vascular remodeling induced by microgravity exposure. Yet, as the most important branch of vasculatures circulating the heart, the coronary artery has been seldomly studied about its adaptations under microgravity conditions. Large-conductance Ca2+-activated potassium channel (BKCa) and the Ras homolog family member A (RhoA)/Rho kinase (ROCK) pathway play key roles in control of vascular tone and mediation of microgravity-induced vascular adjustments. Therefore, we investigated the adaptation of coronary vasoreactivity to simulated microgravity and the role of BKCa and the RhoA/ROCK pathway in it. Four-week-old hind-limb unweighted (HU) rats were adopted to simulate effects of microgravity. Right coronary artery (RCA) constriction was measured by isometric force recording. The activity and expression of BKCa and the RhoA/ROCK pathway were examined by Western blot, patch-clamp recordings, and immunoprecipitation. We found HU significantly decreased RCA vasoconstriction to KCl, serotonin, and U-46619, but increased protein expression and current densities of BKCa, inhibition of which by iberiotoxin (IBTX) further decreased RCA vasoconstriction (P < 0.05). Expression of RhoA and ROCK as well as active RhoA and phosphorylation of myosin light chain (MLC) at Ser19 and MLC phosphatase target-1 at Thr696 were significantly increased by HU, and ROCK inhibitor Y-27632 exerted greater suppressing effect on HU RCA vasoconstriction than that of control (P < 0.05). BKCa opener NS1619 increased HU RCA vasoconstriction, which was blocked by both RhoA and ROCK inhibitor, similar to the effect of IBTX. These results indicate that HU impairs coronary vasoconstriction but enhances BKCa activity acting as a protective mechanism avoiding excessive decrease of coronary vasoreactivity through activation of the RhoA/ROCK pathway.-Wu, Y., Yue, Z., Wang, Q., Lv, Q., Liu, H., Bai, Y., Li, S., Xie, M., Bao, J., Ma, J., Zhu, X., Wang, Z. BKCa compensates impaired coronary vasoreactivity through RhoA/ROCK pathway in hind-limb unweighted rats.
Collapse
Affiliation(s)
- Yue Wu
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China.,Department of Medical Administration, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhijie Yue
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China.,Department of Cardiology, Affiliated Hospital of The Bethune Medical Noncommissioned Officer (NCO) School, Army Medical University, Shijiazhuang, China
| | - Qiguang Wang
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Qiang Lv
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Huan Liu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Yungang Bai
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Shaohua Li
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Manjiang Xie
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Junxiang Bao
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Xianyang Zhu
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China
| | - Zhongchao Wang
- Department of Congenital Heart Disease, General Hospital of Northern Theater Command, Shenyang, China.,Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
63
|
Tahimic CGT, Paul AM, Schreurs AS, Torres SM, Rubinstein L, Steczina S, Lowe M, Bhattacharya S, Alwood JS, Ronca AE, Globus RK. Influence of Social Isolation During Prolonged Simulated Weightlessness by Hindlimb Unloading. Front Physiol 2019; 10:1147. [PMID: 31572207 PMCID: PMC6753329 DOI: 10.3389/fphys.2019.01147] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 08/22/2019] [Indexed: 12/15/2022] Open
Abstract
The hindlimb unloading (HU) model has been used extensively to simulate the cephalad fluid shift and musculoskeletal disuse observed in spaceflight with its application expanding to study immune, cardiovascular and central nervous system responses, among others. Most HU studies are performed with singly housed animals, although social isolation also can substantially impact behavior and physiology, and therefore may confound HU experimental results. Other HU variants that allow for paired housing have been developed although no systematic assessment has been made to understand the effects of social isolation on HU outcomes. Hence, we aimed to determine the contribution of social isolation to tissue responses to HU. To accomplish this, we developed a refinement to the traditional NASA Ames single housing HU system to accommodate social housing in pairs, retaining desirable features of the original design. We conducted a 30-day HU experiment with adult, female mice that were either singly or socially housed. HU animals in both single and social housing displayed expected musculoskeletal deficits versus housing matched, normally loaded (NL) controls. However, select immune and hypothalamic-pituitary-adrenal (HPA) axis responses were differentially impacted by the HU social environment relative to matched NL controls. HU led to a reduction in % CD4+ T cells in singly housed, but not in socially housed mice. Unexpectedly, HU increased adrenal gland mass in socially housed but not singly housed mice, while social isolation increased adrenal gland mass in NL controls. HU also led to elevated plasma corticosterone levels at day 30 in both singly and socially housed mice. Thus, musculoskeletal responses to simulated weightlessness are similar regardless of social environment with a few differences in adrenal and immune responses. Our findings show that combined stressors can mask, not only exacerbate, select responses to HU. These findings further expand the utility of the HU model for studying possible combined effects of spaceflight stressors.
Collapse
Affiliation(s)
- Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,KBR, Houston, TX, United States
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Universities Space Research Association, Columbia, MD, United States
| | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,KBR, Houston, TX, United States
| | - Samantha M Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Blue Marble Space Institute of Science, Seattle, WA, United States
| | - Linda Rubinstein
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Universities Space Research Association, Columbia, MD, United States
| | - Sonette Steczina
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Blue Marble Space Institute of Science, Seattle, WA, United States
| | - Moniece Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Blue Marble Space Institute of Science, Seattle, WA, United States
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| | - April E Ronca
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States.,Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, United States
| |
Collapse
|
64
|
Kravtsova VV, Saburova EA, Krivoi II. The Structural and Functional Characteristics of Rat Soleus Endplates under Short-Term Disruption of Motor Activity. Biophysics (Nagoya-shi) 2019. [DOI: 10.1134/s0006350919050129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
65
|
Usik MA, Ogneva IV. DNA Methylation in Mouse Spermatozoa under Long-Term Modeling the Effects of Microgravity. Russ J Dev Biol 2019. [DOI: 10.1134/s1062360419040076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
66
|
Durand M, Collombet JM, Frasca S, Sarilar V, Lataillade JJ, Le Bousse-Kerdilès MC, Holy X. Separate and combined effects of hypobaric hypoxia and hindlimb suspension on skeletal homeostasis and hematopoiesis in mice. HYPOXIA 2019; 7:41-52. [PMID: 31440522 PMCID: PMC6667353 DOI: 10.2147/hp.s195827] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/15/2019] [Indexed: 12/14/2022]
Abstract
Purpose Bone marrow response to an organismal stress is made by orchestrating the interplay between hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs). Neither the cellular nor the molecular factors that regulate this process are fully understood, especially since this mechanism probably varies depending on the type of stress. Herein, we explored the differentiation and fate of MSCs and HSPCs in mice challenged with a hematopoietic stress or a mechanical stress applied separately or in combination. Methods Mice were subjected to 4 days of hypobaric hypoxia (hematopoietic challenge) and/or 7 days of hindlimb suspension (stromal challenge) and then sacrificed for blood and bone collection. Using hematological measurements, colony-forming unit assays, bone histomorphometry and array-based multiplex ELISA analysis, we evaluated challenge influences on both MSC and HSPC mobilization, differentiation (osteoblasts, osteoclasts, and mature blood cells) and fate. Results We found that hypoxia leads to HSPC mobilization and that an imbalance between bone formation and bone resorption accounts for this mobilization. Whilst suspension is also associated with an imbalance between bone formation and bone resorption, it does not induce HSPC mobilization. Then, we revealed cellular interactions by combining hematopoietic and stromal challenges together in mice. We showed that the hypoxia-driven HSPC mobilization is moderated by suspension. Moreover, when applied in a hypoxic environment, suspension offsets bone imbalance. We identified stroma cell-derived factors MIP-1α, HGF and SDF-1 as potent molecular key players sustaining interactions between hindlimb suspension and hypobaric hypoxia. Conclusion Taken together, our data highlight the benefit of combining different types of stress to better understand the interplay between MSCs and HSPCs.
Collapse
Affiliation(s)
- Marjorie Durand
- Department of Medical and Surgical Assistance to the Armed Forces, French Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex 91223, France
| | - Jean-Marc Collombet
- Department of Medical and Surgical Assistance to the Armed Forces, French Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex 91223, France
| | - Sophie Frasca
- Department of Medical and Surgical Assistance to the Armed Forces, French Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex 91223, France
| | - Véronique Sarilar
- Department of Platforms & Technological Research, French Armed Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex, 91223, France
| | - Jean-Jacques Lataillade
- Department of Medical and Surgical Assistance to the Armed Forces, French Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex 91223, France.,Unit for Research Development, Armed Forces Blood Transfusion Center, Clamart, Cedex 92141, France
| | | | - Xavier Holy
- Department of Platforms & Technological Research, French Armed Forces Biomedical Research Institute (IRBA), Brétigny sur Orge, Cedex, 91223, France
| |
Collapse
|
67
|
Longitudinal time course of muscle impairments during partial weight-bearing in rats. NPJ Microgravity 2019; 5:20. [PMID: 31453318 PMCID: PMC6706399 DOI: 10.1038/s41526-019-0080-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 07/31/2019] [Indexed: 01/14/2023] Open
Abstract
In the near future, space agencies plan to send the first crews for extended stays on the Moon and Mars, where gravity is significantly reduced compared to Earth (0.16×g and 0.38×g, respectively). However, the long-term effects of partial gravity have not yet been elucidated, and ensuring astronauts' health and performance is crucial to the success of these missions. Using a quadrupedal partial weight-bearing (PWB) model in rats that we designed, we investigated the longitudinal time course of muscle function at three different PWB levels. We demonstrated that both muscle mass and muscle function are significantly impaired in reduced weight-bearing environments as early as after 7 days of suspension. Moreover, we showed that muscular alterations are correlated to the PWB level and do not reach a plateau during a 1-month exposure to reduced weight-bearing, emphasizing the need for mitigating countermeasures for safe and successful extraterrestrial exploration.
Collapse
|
68
|
Overbey EG, Paul AM, da Silveira WA, Tahimic CGT, Reinsch SS, Szewczyk N, Stanbouly S, Wang C, Galazka JM, Mao XW. Mice Exposed to Combined Chronic Low-Dose Irradiation and Modeled Microgravity Develop Long-Term Neurological Sequelae. Int J Mol Sci 2019; 20:ijms20174094. [PMID: 31443374 PMCID: PMC6747492 DOI: 10.3390/ijms20174094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023] Open
Abstract
Spaceflight poses many challenges for humans. Ground-based analogs typically focus on single parameters of spaceflight and their associated acute effects. This study assesses the long-term transcriptional effects following single and combination spaceflight analog conditions using the mouse model: simulated microgravity via hindlimb unloading (HLU) and/or low-dose γ-ray irradiation (LDR) for 21 days, followed by 4 months of readaptation. Changes in gene expression and epigenetic modifications in brain samples during readaptation were analyzed by whole transcriptome shotgun sequencing (RNA-seq) and reduced representation bisulfite sequencing (RRBS). The results showed minimal gene expression and cytosine methylation alterations at 4 months readaptation within single treatment conditions of HLU or LDR. In contrast, following combined HLU+LDR, gene expression and promoter methylation analyses showed multiple altered pathways involved in neurogenesis and neuroplasticity, the regulation of neuropeptides, and cellular signaling. In brief, neurological readaptation following combined chronic LDR and HLU is a dynamic process that involves pathways that regulate neuronal function and structure and may lead to late onset neurological sequelae.
Collapse
Affiliation(s)
- Eliah G Overbey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Universities Space Research Association, Columbia, MD 21046, USA
| | - Willian A da Silveira
- Institute for Global Food Security (IGF), School of Biological Sciences, Queen's University, Belfast, Northern Ireland BT7 1NN, UK
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- KBR, Moffett Field, CA 94035, USA
| | - Sigrid S Reinsch
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Nathaniel Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research & National Institute for Health Research Nottingham Biomedical Research Centre, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK
| | - Seta Stanbouly
- Division of Biomedical Engineering Sciences (BMES), Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA
| | - Charles Wang
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jonathan M Galazka
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| | - Xiao Wen Mao
- Division of Biomedical Engineering Sciences (BMES), Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
69
|
Mortreux M, Riveros D, Bouxsein ML, Rutkove SB. A Moderate Daily Dose of Resveratrol Mitigates Muscle Deconditioning in a Martian Gravity Analog. Front Physiol 2019; 10:899. [PMID: 31379604 PMCID: PMC6656861 DOI: 10.3389/fphys.2019.00899] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/27/2019] [Indexed: 11/22/2022] Open
Abstract
While there is a relatively good understanding of the effects of microgravity on human physiology based on five decades of experience, the physiological consequences of partial gravity remain far less well understood. Until recently, no model had been able to replicate partial gravity such as that experienced on Mars (0.38 g), which would be critical to help sustain long-term missions and ensure a safe return to Earth. Recent development of two partial weight bearing (PWB) models, one in mice and one in rats, now allows for quadrupedal partial unloading that mimics Martian gravity. Resveratrol (RSV), a polyphenol most commonly found in grapes and blueberries, has been extensively investigated for its health benefits, including its anti-inflammatory, anti-oxidative, and anti-diabetic effects. In the context of mechanical unloading, RSV has also been shown to preserve bone and muscle mass. However, there is a lack of research regarding its effect on the musculoskeletal system in partial gravity. We hypothesized that a moderate daily dose of RSV (150 mg/kg/day) would help mitigate muscle deconditioning in a Mars gravity analog. Indeed, our results demonstrate that RSV treatment during partial unloading significantly preserves muscle function (e.g., the average change in grip force after 14 days of PWB40 was of −6.18, and +10.92% when RSV was administered) and mitigates muscle atrophy (e.g., RSV supplementation led to an increase of 21.6% in soleus weight for the unloaded animals). This work suggests the potential of a nutraceutical approach to reduce musculoskeletal deconditioning on a long-term mission to Mars.
Collapse
Affiliation(s)
- Marie Mortreux
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Daniela Riveros
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Mary L Bouxsein
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
70
|
DNA Methylation of Mouse Testes, Cardiac and Lung Tissue During Long-Term Microgravity Simulation. Sci Rep 2019; 9:7974. [PMID: 31138883 PMCID: PMC6538624 DOI: 10.1038/s41598-019-44468-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 05/17/2019] [Indexed: 01/05/2023] Open
Abstract
Under microgravity, the gene expression levels vary in different types of cells; however, the reasons for this have not been sufficiently studied. The aim of this work was to evaluate the methylation of CpG islands in the promoter regions of the genes encoding some cytoskeletal proteins, the total methylation and 5 hmC levels, and the levels of enzymes that regulate these processes in the testes, heart, and lungs in mice after a 30-day microgravity modeling by antiorthostatic suspension and after a subsequent 12-hour recovery as well as in the corresponding control group and identical groups treated with essential phospholipids. The obtained results indicate that under modeling microgravity in the examined tissues a decrease of cytoskeletal gene expression (mainly in the heart and lungs tissues) correlated with an increase in the CpG islands methylation and an increase of the expression (mainly in the testes tissue) - with a decrease of the CpG-methylation, despite of the fact that in the examined tissues took place a decrease of the content methylases and demethylases. But the deacetylase HDAC1 content increased in the heart and lungs tissues and decreased in the testes, letting us suggest its participation in the regulation of the methylation level under microgravity conditions.
Collapse
|
71
|
Song H, Cho S, Lee HY, Lee H, Song W. The Effects of Progressive Resistance Exercise on Recovery Rate of Bone and Muscle in a Rodent Model of Hindlimb Suspension. Front Physiol 2018; 9:1085. [PMID: 30150940 PMCID: PMC6099092 DOI: 10.3389/fphys.2018.01085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/23/2018] [Indexed: 11/28/2022] Open
Abstract
Purpose: This study aimed to examine the exercise-mediated musculoskeletal recovery following hindlimb suspension (HS) in order to identify whether bone modeling and muscle hypertrophy would eventuate in a synchronized manner during recovery stage. Methods: To identify whether 2-week HS would be sufficient to induce a significant reduction of physiological indices in both tibia and adjacent hindlimb muscles, a total of 20 rats was randomized into 2-week HS (n = 10) and age-matched control group (n = 10, CON). Another batch of rats were randomly assigned to three different groups to identify recovery intervention effects following suspension: (1) 2-week HS followed by 4-week spontaneous reloading recovery (HRE, n = 7). (2) 2-week HS followed by 4-week progressive resistance ladder climbing exercise (HEX, n = 7). (3) Age-matched control (CON, n = 7). DXA, micro-CT, and 18F-sodium fluoride (NaF) imaging, and EIA analysis were utilized to measure tibia bone indices. Hindlimb muscle wet weight and grip strength were measured to evaluate muscle mass and strength, respectively. Results: In study 1, bone quality values [bone volume/total volume (BV/TV): -27%, areal bone mineral density (aBMD): -23%, mineral contents: -7.9%, mineral density: –4.1%, and bone density: -38.9%] and skeletal muscle weight (soleus: -46.8%, gastrocnemius: -19.6%, plantaris: -20.8%, TA: -22.8%, and EDL: -9.9%) were significantly lower in HS group compared to CON group. In study 2, micro-CT and DXA-based bone morphology (bone density, BT/TV, and aBMD) were fully recovered in HRE or HEX group. However, suspension-induced dysregulation of bone mineral metabolism was returned to age-matched control group in only HEX group, but not in HRE group. A greater level of biomarkers of bone formation (P1NF) and resorption (CTX-1) was observed in only HRE group compared to CON. The hindlimb skeletal muscle mass was significantly lower in both HRE and HEX groups compared to CON group. Hindlimb grip strength was the greatest in HEX group, followed by CON and HRE groups. Conclusion: Following HS, progressive resistance exercise promotes recovery rates of bone and skeletal muscle strength without a significant increase in muscular mass, suggesting that exercise-induced reacquisition of bone and muscle strength is independent of muscle hypertrophy during early recovery stage.
Collapse
Affiliation(s)
- Hansol Song
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Suhan Cho
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, South Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hojun Lee
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Sports and Health Science, Kyungsung University, Busan, South Korea
| | - Wook Song
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul National University, Seoul, South Korea.,Institute on Aging, Seoul National University, Seoul, South Korea
| |
Collapse
|
72
|
Shimkus KL, Shirazi-Fard Y, Wiggs MP, Ullah ST, Pohlenz C, Gatlin DM, Carroll CC, Hogan HA, Fluckey JD. Responses of skeletal muscle size and anabolism are reproducible with multiple periods of unloading/reloading. J Appl Physiol (1985) 2018; 125:1456-1467. [PMID: 30091665 DOI: 10.1152/japplphysiol.00736.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanical unloading has long been understood to contribute to rapid and substantial adaptations within skeletal muscle, most notably, muscle atrophy. Studies have often demonstrated that many of the alterations resulting from disuse are reversed with a reintroduction of load and have supported the concept of muscle plasticity. We hypothesized that adaptations during disuse and recovery were a repeatable/reproducible phenomenon, which we tested with repeated changes in mechanical load. Rats were assigned to one of the following five groups: animals undergoing one or two bouts of hindlimb unloading (28 days), with or without recovery (56 day), or control. Following the completion of their final time point, posterior crural muscles were studied. Muscle sizes were lower following 28 days of disuse but fully recovered with a 56-day reloading period, regardless of the number of disuse/recovery cycles. Mixed protein fractional synthesis rates consistently reflected mass and loading conditions (supported by anabolic signaling), whereas the myofibrillar protein synthesis response varied among muscles. Amino acid concentrations were assessed in the gastrocnemius free pool and did not correlate with muscle atrophy associated with mechanical unloading. Muscle collagen concentrations were higher following the second unloading period and remained elevated following 56 days of recovery. Anabolic responses to alterations in load are preserved throughout multiple perturbations, but repeated periods of unloading may cause additive strain to muscle structure (collagen). This study suggests that whereas mass and anabolism are reproducibly reflective of the loading environment, repeated exposure to unloading and/or reloading may impact the overall structural integrity of muscle. NEW & NOTEWORTHY Repeatability should be considered a component of skeletal muscle plasticity during atrophy and recovery. Muscle anabolism is equally affected during a first or second disuse bout and returns equally with adequate recovery. Elevated muscle collagen concentrations observed after the second unloading period suggest altered structural integrity with repeated disuse.
Collapse
Affiliation(s)
- Kevin L Shimkus
- Department of Health and Kinesiology, Texas A&M University , College Station, Texas
| | - Yasaman Shirazi-Fard
- Department of Mechanical Engineering, Texas A&M University , College Station, Texas
| | - Michael P Wiggs
- Department of Health and Kinesiology, Texas A&M University , College Station, Texas
| | - Shaik T Ullah
- Department of Health and Kinesiology, Texas A&M University , College Station, Texas
| | - Camilo Pohlenz
- Department of Wildlife and Fisheries, Texas A&M University , College Station, Texas
| | - Delbert M Gatlin
- Department of Wildlife and Fisheries, Texas A&M University , College Station, Texas
| | - Chad C Carroll
- Department of Health and Kinesiology, Purdue University , West Lafayette, Indiana
| | - Harry A Hogan
- Department of Mechanical Engineering, Texas A&M University , College Station, Texas
| | - James D Fluckey
- Department of Health and Kinesiology, Texas A&M University , College Station, Texas
| |
Collapse
|
73
|
Chibalin AV, Benziane B, Zakyrjanova GF, Kravtsova VV, Krivoi II. Early endplate remodeling and skeletal muscle signaling events following rat hindlimb suspension. J Cell Physiol 2018; 233:6329-6336. [PMID: 29719042 DOI: 10.1002/jcp.26594] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 03/12/2018] [Indexed: 12/30/2022]
Abstract
Motor endplates naturally undergo continual morphological changes that are altered in response to changes in neuromuscular activity. This study examines the consequences of acute (6-12 hr) disuse following hindlimb suspension on rat soleus muscle endplate structural stability. We identify early changes in several key signaling events including markers of protein kinase activation, AMPK phosphorylation and autophagy markers which may play a role in endplate remodeling. Acute disuse does not change endplate fragmentation, however, it decreases both the individual fragments and the total endplate area. This decrease was accompanied by an increase in the mean fluorescence intensity from the nicotinic acetylcholine receptors which compensate the endplate area loss. Muscle disuse decreased phosphorylation of AMPK and its substrate ACC, and stimulated mTOR controlled protein synthesis pathway and stimulated autophagy. Our findings provide evidence that changes in endplate stability are accompanied by reduced AMPK phosphorylation and an increase in autophagy markers, and these changes are evident within hours of onset of skeletal muscle disuse.
Collapse
Affiliation(s)
- Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Boubacar Benziane
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Guzalija F Zakyrjanova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia.,Department of Normal Physiology, Kazan State Medical University, Kazan, Russia
| | - Violetta V Kravtsova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Igor I Krivoi
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
74
|
Mortreux M, Nagy JA, Ko FC, Bouxsein ML, Rutkove SB. A novel partial gravity ground-based analog for rats via quadrupedal unloading. J Appl Physiol (1985) 2018; 125:175-182. [PMID: 29565773 DOI: 10.1152/japplphysiol.01083.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Musculoskeletal deconditioning is a well-known consequence of microgravity. However, the effects of partial gravity, such as that experienced on the moon (0.16 g) or Mars (0.38 g), on musculoskeletal health remain relatively unexplored. Because Mars is being increasingly viewed as the likely next extraterrestrial site for human exploration, there is an increasing need for Earth-based models that can replicate the long-term physiological effects of microgravity. These models would also offer the opportunity to explore the potential impact of partial artificial gravity (as would be achieved by centrifugation). In this study, we describe a novel partial gravity model that can be employed in rats over extended periods of time. We demonstrate that 2 wk of partial weight bearing at 20, 40, or 70% of normal loading affects the musculoskeletal health of the animals, as evidenced by decreased trabecular bone density (ranging from -7.5 ± 2.7% at 70% of normal loading to -27.9 ± 2.9% at 20%), hindlimb muscle mass, and impaired muscle function as characterized by grip force. This new model will facilitate studies of the physiological changes occurring in partial gravity and allow for the design of potential countermeasures to mitigate these changes. NEW & NOTEWORTHY This research article describes the first quadrupedal unloading model in rats that is sustainable for investigating the physiological alterations occurring in partial gravity environments, providing a new and adaptable model for ground-based research for future space exploration.
Collapse
Affiliation(s)
- Marie Mortreux
- Department of Neurology, Harvard Medical School-Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Janice A Nagy
- Department of Neurology, Harvard Medical School-Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Frank C Ko
- Department of Orthopedics, Harvard Medical School-Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Mary L Bouxsein
- Department of Orthopedics, Harvard Medical School-Beth Israel Deaconess Medical Center , Boston, Massachusetts
| | - Seward B Rutkove
- Department of Neurology, Harvard Medical School-Beth Israel Deaconess Medical Center , Boston, Massachusetts
| |
Collapse
|
75
|
Jiang M, Lyu Q, Bai YG, Liu H, Yang J, Cheng JH, Zheng M, Ma J. Focal adhesions are involved in simulated-microgravity-induced basilar and femoral arterial remodelling in rats. Can J Physiol Pharmacol 2018. [PMID: 29527943 DOI: 10.1139/cjpp-2017-0665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies have suggested that microgravity-induced arterial remodelling contributes to post-flight orthostatic intolerance and that multiple mechanisms are involved in arterial remodelling. However, the initial mechanism by which haemodynamic changes induce arterial remodelling is unknown. Focal adhesions (FAs) are dynamic protein complexes that have mechanotransduction properties. This study aimed to investigate the role of FAs in simulated-microgravity-induced basilar and femoral arterial remodelling. A 4-week hindlimb-unweighted (HU) rat model was used to simulate the effects of microgravity, and daily 1-hour intermittent artificial gravity (IAG) was used to prevent arterial remodelling. After 4-week HU, wall thickness, volume of smooth muscle cells (SMCs) and collagen content were increased in basilar artery but decreased in femoral artery (P < 0.05). Additionally, the expression of p-FAK Y397 and p-Src Y418 was increased and reduced in SMCs of basilar and femoral arteries, respectively, by HU (P < 0.05). The number of FAs was increased in basilar artery and reduced in femoral artery by HU (P < 0.05). Furthermore, daily 1-hour IAG prevented HU-induced differential structural adaptations and changes in FAs of basilar and femoral arteries. These results suggest that FAs may act as mechanosensors in arterial remodelling by initiating intracellular signal transduction in response to altered mechanical stress induced by microgravity.
Collapse
Affiliation(s)
- Min Jiang
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Qiang Lyu
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yun-Gang Bai
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Huan Liu
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jing Yang
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jiu-Hua Cheng
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Ming Zheng
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jin Ma
- Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aerospace Physiology, Key Laboratory of Aerospace Medicine of Ministry of Education, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
76
|
|
77
|
Childress P, Brinker A, Gong CMS, Harris J, Olivos DJ, Rytlewski JD, Scofield DC, Choi SY, Shirazi-Fard Y, McKinley TO, Chu TMG, Conley CL, Chakraborty N, Hammamieh R, Kacena MA. Forces associated with launch into space do not impact bone fracture healing. LIFE SCIENCES IN SPACE RESEARCH 2018; 16:52-62. [PMID: 29475520 PMCID: PMC5828031 DOI: 10.1016/j.lssr.2017.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 06/08/2023]
Abstract
Segmental bone defects (SBDs) secondary to trauma invariably result in a prolonged recovery with an extended period of limited weight bearing on the affected limb. Soldiers sustaining blast injuries and civilians sustaining high energy trauma typify such a clinical scenario. These patients frequently sustain composite injuries with SBDs in concert with extensive soft tissue damage. For soft tissue injury resolution and skeletal reconstruction a patient may experience limited weight bearing for upwards of 6 months. Many small animal investigations have evaluated interventions for SBDs. While providing foundational information regarding the treatment of bone defects, these models do not simulate limited weight bearing conditions after injury. For example, mice ambulate immediately following anesthetic recovery, and in most cases are normally ambulating within 1-3 days post-surgery. Thus, investigations that combine disuse with bone healing may better test novel bone healing strategies. To remove weight bearing, we have designed a SBD rodent healing study in microgravity (µG) on the International Space Station (ISS) for the Rodent Research-4 (RR-4) Mission, which launched February 19, 2017 on SpaceX CRS-10 (Commercial Resupply Services). In preparation for this mission, we conducted an end-to-end mission simulation consisting of surgical infliction of SBD followed by launch simulation and hindlimb unloading (HLU) studies. In brief, a 2 mm defect was created in the femur of 10 week-old C57BL6/J male mice (n = 9-10/group). Three days after surgery, 6 groups of mice were treated as follows: 1) Vivarium Control (maintained continuously in standard cages); 2) Launch Negative Control (placed in the same spaceflight-like hardware as the Launch Positive Control group but were not subjected to launch simulation conditions); 3) Launch Positive Control (placed in spaceflight-like hardware and also subjected to vibration followed by centrifugation); 4) Launch Positive Experimental (identical to Launch Positive Control group, but placed in qualified spaceflight hardware); 5) Hindlimb Unloaded (HLU, were subjected to HLU immediately after launch simulation tests to simulate unloading in spaceflight); and 6) HLU Control (single housed in identical HLU cages but not suspended). Mice were euthanized 28 days after launch simulation and bone healing was examined via micro-Computed Tomography (µCT). These studies demonstrated that the mice post-surgery can tolerate launch conditions. Additionally, forces and vibrations associated with launch did not impact bone healing (p = .3). However, HLU resulted in a 52.5% reduction in total callus volume compared to HLU Controls (p = .0003). Taken together, these findings suggest that mice having a femoral SBD surgery tolerated the vibration and hypergravity associated with launch, and that launch simulation itself did not impact bone healing, but that the prolonged lack of weight bearing associated with HLU did impair bone healing. Based on these findings, we proceeded with testing the efficacy of FDA approved and novel SBD therapies using the unique spaceflight environment as a novel unloading model on SpaceX CRS-10.
Collapse
Affiliation(s)
- Paul Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - Alexander Brinker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - Cynthia-May S Gong
- KBR Wyle Laboratory and Division of Space Biology, NASA Ames Research Center, Moffett Field, CA, United States
| | - Jonathan Harris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - David J Olivos
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - Jeffrey D Rytlewski
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - David C Scofield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - Sungshin Y Choi
- KBR Wyle Laboratory and Division of Space Biology, NASA Ames Research Center, Moffett Field, CA, United States
| | - Yasaman Shirazi-Fard
- KBR Wyle Laboratory and Division of Space Biology, NASA Ames Research Center, Moffett Field, CA, United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States
| | - Tien-Min G Chu
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, United States
| | - Carolynn L Conley
- Department of Defense Space Test Program, Houston, TX, United States
| | - Nabarun Chakraborty
- Geneva Foundation, Fredrick, MD, United States; US Army Center for Environmental Health Research, Fredrick, MD, United States
| | - Rasha Hammamieh
- US Army Center for Environmental Health Research, Fredrick, MD, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, United States.
| |
Collapse
|
78
|
Radugina EA, Almeida EAC, Blaber E, Poplinskaya VA, Markitantova YV, Grigoryan EN. Exposure to microgravity for 30 days onboard Bion M1 caused muscle atrophy and impaired regeneration in murine femoral Quadriceps. LIFE SCIENCES IN SPACE RESEARCH 2018; 16:18-25. [PMID: 29475516 DOI: 10.1016/j.lssr.2017.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/25/2017] [Accepted: 08/18/2017] [Indexed: 06/08/2023]
Abstract
Mechanical unloading in microgravity during spaceflight is known to cause muscular atrophy, changes in muscle fiber composition, gene expression, and reduction in regenerative muscle growth. Although some limited data exists for long-term effects of microgravity in human muscle, these processes have mostly been studied in rodents for short periods of time. Here we report on how long-term (30-day long) mechanical unloading in microgravity affects murine muscles of the femoral Quadriceps group. To conduct these studies we used muscle tissue from 6 microgravity mice, in comparison to habitat (7), and vivarium (14) ground control mice from the NASA Biospecimen Sharing Program conducted in collaboration with the Institute for Biomedical Problems of the Russian Academy of Sciences, during the Russian Bion M1 biosatellite mission in 2013. Muscle histomorphology from microgravity specimens showed signs of extensive atrophy and regenerative hypoplasia relative to ground controls. Specifically, we observed a two-fold decrease in the number of myonuclei, compared to vivarium and ground controls, and central location of myonuclei, low density of myofibers in the tissue, and of myofibrils within a fiber, as well as fragmentation and swelling of myofibers. Despite obvious atrophy, muscle regeneration nevertheless appeared to have continued after 30 days in microgravity as evidenced by thin and short newly formed myofibers. Many of them, however, showed evidence of apoptotic cells and myofibril degradation, suggesting that long-term unloading in microgravity may affect late stages of myofiber differentiation. Ground asynchronous and vivarium control animals demonstrated normal, well-developed tissue structure with sufficient blood and nerve supply and evidence of regenerative formation of new myofibers free of apoptotic nuclei. Regenerative activity of satellite cells in muscles was observed both in microgravity and ground control groups, using Pax7 and Myogenin immunolocalization, as well as Myogenin expression analysis. In addition, we have detected positive nuclear immunolocalization of c-Jun and c-Myc proteins indicating their sensitivity to changes in gravitational loading in a given model. In summary, long-term spaceflight in microgravity caused significant atrophy and degeneration of the femoral Quadriceps muscle group, and it may interfere with muscle regenerative processes by inducing apoptosis in newly-formed myofibrils during their differentiation phase.
Collapse
Affiliation(s)
- E A Radugina
- Koltzov Institute of Developmental Biology RAS, 26 Vavilova street, Moscow 119334, Russia.
| | - E A C Almeida
- NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - E Blaber
- NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - V A Poplinskaya
- Koltzov Institute of Developmental Biology RAS, 26 Vavilova street, Moscow 119334, Russia
| | - Y V Markitantova
- Koltzov Institute of Developmental Biology RAS, 26 Vavilova street, Moscow 119334, Russia
| | - E N Grigoryan
- Koltzov Institute of Developmental Biology RAS, 26 Vavilova street, Moscow 119334, Russia
| |
Collapse
|
79
|
Zhang LF, Hargens AR. Spaceflight-Induced Intracranial Hypertension and Visual Impairment: Pathophysiology and Countermeasures. Physiol Rev 2017; 98:59-87. [PMID: 29167331 DOI: 10.1152/physrev.00017.2016] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
Visual impairment intracranial pressure (VIIP) syndrome is considered an unexplained major risk for future long-duration spaceflight. NASA recently redefined this syndrome as Spaceflight-Associated Neuro-ocular Syndrome (SANS). Evidence thus reviewed supports that chronic, mildly elevated intracranial pressure (ICP) in space (as opposed to more variable ICP with posture and activity on Earth) is largely accounted for by loss of hydrostatic pressures and altered hemodynamics in the intracranial circulation and the cerebrospinal fluid system. In space, an elevated pressure gradient across the lamina cribrosa, caused by a chronic but mildly elevated ICP, likely elicits adaptations of multiple structures and fluid systems in the eye which manifest themselves as the VIIP syndrome. A chronic mismatch between ICP and intraocular pressure (IOP) in space may acclimate the optic nerve head, lamina cribrosa, and optic nerve subarachnoid space to a condition that is maladaptive to Earth, all contributing to the pathogenesis of space VIIP syndrome. Relevant findings help to evaluate whether artificial gravity is an appropriate countermeasure to prevent this seemingly adverse effect of long-duration spaceflight.
Collapse
Affiliation(s)
- Li-Fan Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China; and Department of Orthopaedic Surgery, University of California, San Diego, California
| | - Alan R Hargens
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China; and Department of Orthopaedic Surgery, University of California, San Diego, California
| |
Collapse
|
80
|
Yu K, Doherty AH, Genik PC, Gookin SE, Roteliuk DM, Wojda SJ, Jiang ZS, McGee-Lawrence ME, Weil MM, Donahue SW. Mimicking the effects of spaceflight on bone: Combined effects of disuse and chronic low-dose rate radiation exposure on bone mass in mice. LIFE SCIENCES IN SPACE RESEARCH 2017; 15:62-68. [PMID: 29198315 DOI: 10.1016/j.lssr.2017.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 06/07/2023]
Abstract
During spaceflight, crewmembers are subjected to biomechanical and biological challenges including microgravity and radiation. In the skeleton, spaceflight leads to bone loss, increasing the risk of fracture. Studies utilizing hindlimb suspension (HLS) as a ground-based model of spaceflight often neglect the concomitant effects of radiation exposure, and even when radiation is accounted for, it is often delivered at a high-dose rate over a very short period of time, which does not faithfully mimic spaceflight conditions. This study was designed to investigate the skeletal effects of low-dose rate gamma irradiation (8.5 cGy gamma radiation per day for 20 days, amounting to a total dose of 1.7 Gy) when administered simultaneously to disuse from HLS. The goal was to determine whether continuous, low-dose rate radiation administered during disuse would exacerbate bone loss in a murine HLS model. Four groups of 16 week old female C57BL/6 mice were studied: weight bearing + no radiation (WB+NR), HLS + NR, WB + radiation exposure (WB+RAD), and HLS+RAD. Surprisingly, although HLS led to cortical and trabecular bone loss, concurrent radiation exposure did not exacerbate these effects. Our results raise the possibility that mechanical unloading has larger effects on the bone loss that occurs during spaceflight than low-dose rate radiation.
Collapse
Affiliation(s)
- Kanglun Yu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Alison H Doherty
- Department of Medical Education, WWAMI Medical Education Program, University of Wyoming, Laramie, WY, USA
| | - Paula C Genik
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Sara E Gookin
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Danielle M Roteliuk
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Samantha J Wojda
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Michael M Weil
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Seth W Donahue
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
81
|
Serum miRNAs miR-23a, 206, and 499 as Potential Biomarkers for Skeletal Muscle Atrophy. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8361237. [PMID: 29214178 PMCID: PMC5682897 DOI: 10.1155/2017/8361237] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/26/2017] [Accepted: 07/10/2017] [Indexed: 12/12/2022]
Abstract
Muscle biopsy has long been expected to be replaced by noninvasive biomarkers with diagnostic value and prognostic applications for muscle atrophy. Growing evidence suggests that circulating microRNAs (miRNAs) could act as biomarkers for numerous pathophysiological statuses. In the present study, our results showed that the serum levels of six muscle-specific miRNAs (miR-1/23a/133/206/208b/499) were all elevated in unloading induced mice. The medium levels of these six muscle-specific miRNAs were all elevated in starvation induced atrophic C2C12 myotubes. Moreover, the serum levels of miR-23a/206/499 were induced in participants after 45 days of head-down bed rest (HDBR). The levels of miR-23a/206/499 were positively correlated with the ratio of soleus volume loss in HDBR participants, indicating that they might represent the process of muscle loss. In conclusion, our results demonstrated that circulating miRNAs could serve as useful biochemical and molecular indicators for muscle atrophy diagnosis and disease progression.
Collapse
|
82
|
Colaianni G, Cinti S, Colucci S, Grano M. Irisin and musculoskeletal health. Ann N Y Acad Sci 2017; 1402:5-9. [PMID: 28437576 DOI: 10.1111/nyas.13345] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 01/02/2023]
Abstract
Irisin is a hormone-like myokine produced in abundance by skeletal muscle in response to exercise, both in mice and humans. Once released into the circulation, irisin acts on white adipocytes to induce the browning response and subsequently activates nonshivering thermogenesis. We have examined the premise that irisin produced during exercise may subserve further functions in the musculoskeletal system. We review evidence for its possible skeletal effects, including the central role that irisin plays in the control of bone mass, with positive effects on cortical mineral density and geometry in mice. We also review the autocrine effects of irisin in skeletal muscle, in which it upregulates the expression of its precursor (FNDC5). Since loss of bone and muscle mass occurs with aging, immobility, and several metabolic diseases, future studies exploring the efficacy of irisin in restoring bone and reversing muscle wasting could be important to establishing irisin as a molecule that combines beneficial effects for treating osteoporosis and muscular atrophy. If the results from mice were confirmed in human studies, an irisin-based therapy could be developed for physically disabled or bedridden patients.
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona, Ancona, Italy
| | - Silvia Colucci
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
83
|
Seawright JW, Samman Y, Sridharan V, Mao XW, Cao M, Singh P, Melnyk S, Koturbash I, Nelson GA, Hauer-Jensen M, Boerma M. Effects of low-dose rate γ-irradiation combined with simulated microgravity on markers of oxidative stress, DNA methylation potential, and remodeling in the mouse heart. PLoS One 2017; 12:e0180594. [PMID: 28678877 PMCID: PMC5498037 DOI: 10.1371/journal.pone.0180594] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 06/16/2017] [Indexed: 01/31/2023] Open
Abstract
Purpose Space travel is associated with an exposure to low-dose rate ionizing radiation and the microgravity environment, both of which may lead to impairments in cardiac function. We used a mouse model to determine short- and long-term cardiac effects to simulated microgravity (hindlimb unloading; HU), continuous low-dose rate γ-irradiation, or a combination of HU and low-dose rate γ-irradiation. Methods Cardiac tissue was obtained from female, C57BL/6J mice 7 days, 1 month, 4 months, and 9 months following the completion of a 21 day exposure to HU or a 21 day exposure to low-dose rate γ-irradiation (average dose rate of 0.01 cGy/h to a total of 0.04 Gy), or a 21 day simultaneous exposure to HU and low-dose rate γ-irradiation. Immunoblot analysis, rt-PCR, high-performance liquid chromatography, and histology were used to assess inflammatory cell infiltration, cardiac remodeling, oxidative stress, and the methylation potential of cardiac tissue in 3 to 6 animals per group. Results The combination of HU and γ-irradiation demonstrated the strongest increase in reduced to oxidized glutathione ratios 7 days and 1 month after treatment, but a difference was no longer apparent after 9 months. On the other hand, no significant changes in 4-hydroxynonenal adducts was seen in any of the groups, at the measured endpoints. While manganese superoxide dismutase protein levels decreased 9 months after low-dose γ-radiation, no changes were observed in expression of catalase or Nrf2, a transcription factor that determines the expression of several antioxidant enzymes, at the measured endpoints. Inflammatory marker, CD-2 protein content was significantly decreased in all groups 4 months after treatment. No significant differences were observed in α-smooth muscle cell actin protein content, collagen type III protein content or % total collagen. Conclusions This study has provided the first and relatively broad analysis of small molecule and protein markers of oxidative stress, T-lymphocyte infiltration, and cardiac remodeling in response to HU with simultaneous exposure to low-dose rate γ-radiation. Results from the late observation time points suggest that the hearts had mostly recovered from these two experimental conditions. However, further research is needed with larger numbers of animals for a more robust statistical power to fully characterize the early and late effects of simulated microgravity combined with exposure to low-dose rate ionizing radiation on the heart.
Collapse
Affiliation(s)
- John W. Seawright
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
- * E-mail:
| | - Yusra Samman
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Xiao Wen Mao
- Department of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, The United States of America
| | - Maohua Cao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Preeti Singh
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Stepan Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Gregory A. Nelson
- Department of Basic Sciences and Radiation Medicine, Loma Linda University, Loma Linda, CA, The United States of America
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, The United States of America
| |
Collapse
|
84
|
Anthony SS, Date I, Yasuhara T. Limiting exercise inhibits neuronal recovery from neurological disorders. Brain Circ 2017; 3:124-129. [PMID: 30276313 PMCID: PMC6057693 DOI: 10.4103/bc.bc_16_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/04/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022] Open
Abstract
Patients who are bedridden often suffer from muscular atrophy due to reduced daily activities and can become depressed. However, patients who undergo physical therapy sometimes demonstrate positive benefits including a reduction of stressful and depressed behavior. Regenerative medicine has seen improvements in two stem cell-based therapies for central nervous system disorders. One therapy is through the transfer of exogenous stem cells. The other therapy is a more natural method and focuses on the increasing endogenous neurogenesis and restoring the neurological impairments. This study overviews how immobilization-induced disuse atrophy affects neurogenesis in rats, specifically hypothesizing that immobilization diminishes circulating trophic factor levels, like vascular endothelial growth factors or brain-derived neurotrophic factor, which in turn limits neurogenesis. This hypothesis requires the classification of the stem cell microenvironment by probing growth factors in addition to other stress-related proteins that correlate with exercise-induced neurogenesis. There is research examining the effects of increased exercise on neurogenesis while limiting exercise, which better demonstrates the pathological states of immobile stroke patients, remains relatively unexplored. To examine the effects of immobilization on neurogenesis quantitative measurements of movements, 5-bromo-2deoxyuridine labeling of proliferative cells, biochemical assays of serum, cerebrospinal fluid and neurological levels of trophic factors, growth factors, and stress-related proteins will indicate levels of neurogenesis. In further research, studies are needed to show how in vivo stimulation, or lack thereof, affects stem cell microenvironments to advance treatment procedures for strengthening neurogenesis in bedridden patients. This paper is a review article. Referred literature in this paper has been listed in the references section. The datasets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
- Stefan S. Anthony
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Okayama, Japan
| |
Collapse
|
85
|
Abstract
Drynariae Rhizoma is a kidney-tonifying herb that has a long history in clinical practice for the treatment of bone fractures and joint diseases in China. Flavonoids are considered to be its major active ingredients and are reported to ease bone loss in ovariectomized rats. However, the beneficial effects of the total flavonoids of Drynariae Rhizoma on osteoporosis caused by microgravity or mechanical inactivity remain unknown. This study assessed the effects of total Drynariae Rhizoma flavonoids (DRTF, Qihuang, Beijing, China, national medicine permit No. Z20030007, number of production: 04080081, content of DRTF ≥80%) against bone loss induced by simulated microgravity. A hindlimb unloading tail-suspended rat model was established to determine the effect of DRTF on bone mineral density (BMD), biomechanical strength and trabecular bone microarchitecture. Twenty-eight male Sprague-Dawley rats were divided into four groups: the baseline, control, hindlimb unloading with vehicle (HLU), and hindlimb unloading treated with DRTF (HLU-DRTF, 75 mg/kg/day) groups. Oral DRTF was administered for 4 weeks. The underlying mechanisms of the DRTF actions on disuse-induced osteoporosis are discussed. The results showed that DRTF treatment significantly increased the BMD and mechanical strength of tail-suspended rats. Enhanced bone turnover markers with HLU treatment were attenuated by DRTF administration. Deterioration of trabecular bone induced by HLU was prevented through elevated bone volume/tissue volume (BV/TV), trabecular number (Tb. N), trabecular thickness (Tb. Th) and decreased trabecular separation (Tb. Sp). The present study provides the first evidence that DRTF prevents bone loss induced by HLU treatment, indicating its potential application in the treatment of disuse-induced osteoporosis.
Collapse
|
86
|
Colaianni G, Mongelli T, Cuscito C, Pignataro P, Lippo L, Spiro G, Notarnicola A, Severi I, Passeri G, Mori G, Brunetti G, Moretti B, Tarantino U, Colucci SC, Reseland JE, Vettor R, Cinti S, Grano M. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep 2017; 7:2811. [PMID: 28588307 PMCID: PMC5460172 DOI: 10.1038/s41598-017-02557-8] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 04/13/2017] [Indexed: 01/12/2023] Open
Abstract
We previously showed that Irisin, a myokine released from skeletal muscle after physical exercise, plays a central role in the control of bone mass. Here we report that treatment with recombinant Irisin prevented bone loss in hind-limb suspended mice when administered during suspension (preventive protocol) and induced recovery of bone mass when mice were injected after bone loss due to a suspension period of 4 weeks (curative protocol). MicroCT analysis of femurs showed that r-Irisin preserved both cortical and trabecular bone mineral density, and prevented a dramatic decrease of the trabecular bone volume fraction. Moreover, r-Irisin protected against muscle mass decline in the hind-limb suspended mice, and maintained the fiber cross-sectional area. Notably, the decrease of myosin type II expression in unloaded mice was completely prevented by r-Irisin administration. Our data reveal for the first time that Irisin retrieves disuse-induced bone loss and muscle atrophy. These findings may lead to development of an Irisin-based therapy for elderly immobile osteoporotic and physically disable patients, and might represent a countermeasure for astronauts subjected to microgravity-induced bone and muscle losses.
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Teresa Mongelli
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Concetta Cuscito
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Paolo Pignataro
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Luciana Lippo
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Giovanna Spiro
- Department of Medicine-DIMED, Internal Medicine 3, University of Padova, 35128, Padova, Italy
| | - Angela Notarnicola
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona, 60020, Ancona, Italy
| | - Giovanni Passeri
- Department of Clinical and Experimental Medicine, University of Parma, 43126, Parma, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100, Foggia, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Biagio Moretti
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Umberto Tarantino
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, 00133, Rome, Italy
| | - Silvia C Colucci
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, 70124, Bari, Italy
| | - Janne E Reseland
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Blindern, N-0317, Oslo, Norway
| | - Roberto Vettor
- Department of Medicine-DIMED, Internal Medicine 3, University of Padova, 35128, Padova, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona, 60020, Ancona, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, 70124, Bari, Italy.
| |
Collapse
|
87
|
Bone Marrow Adipose Tissue Deficiency Increases Disuse-Induced Bone Loss in Male Mice. Sci Rep 2017; 7:46325. [PMID: 28402337 PMCID: PMC5389344 DOI: 10.1038/srep46325] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/16/2017] [Indexed: 12/17/2022] Open
Abstract
Bone marrow adipose tissue (MAT) is negatively associated with bone mass. Since osteoblasts and adipocytes are derived from the same precursor cells, adipocyte differentiation may occur at the expense of osteoblast differentiation. We used MAT-deficient KitW/W−v (MAT-) mice to determine if absence of MAT reduced bone loss in hindlimb-unloaded (HU) mice. Male MAT- and wild-type (WT) mice were randomly assigned to a baseline, control or HU group (n = 10 mice/group) within each genotype and HU groups unloaded for 2 weeks. Femurs were evaluated using micro-computed tomography, histomorphometry and targeted gene profiling. MAT- mice had a greater reduction in bone volume fraction after HU than did WT mice. HU MAT- mice had elevated cancellous bone formation and resorption compared to other treatment groups as well as a unique profile of differentially expressed genes. Adoptive transfer of WT bone marrow-derived hematopoietic stem cells reconstituted c-kit but not MAT in KitW/W−v mice. The MAT- WT → KitW/W−v mice lost cancellous bone following 2 weeks of HU. In summary, results from this study suggest that MAT deficiency was not protective, and was associated with exaggerated disuse-induced cancellous bone loss.
Collapse
|
88
|
Petrov AM, Kravtsova VV, Matchkov VV, Vasiliev AN, Zefirov AL, Chibalin AV, Heiny JA, Krivoi II. Membrane lipid rafts are disturbed in the response of rat skeletal muscle to short-term disuse. Am J Physiol Cell Physiol 2017; 312:C627-C637. [PMID: 28274922 DOI: 10.1152/ajpcell.00365.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/22/2017] [Accepted: 03/05/2017] [Indexed: 12/19/2022]
Abstract
Marked loss of skeletal muscle mass occurs under various conditions of disuse, but the molecular and cellular mechanisms leading to atrophy are not completely understood. We investigate early molecular events that might play a role in skeletal muscle remodeling during mechanical unloading (disuse). The effects of acute (6-12 h) hindlimb suspension on the soleus muscles from adult rats were examined. The integrity of plasma membrane lipid rafts was tested utilizing cholera toxin B subunit or fluorescent sterols. In addition, resting intracellular Ca2+ level was analyzed. Acute disuse disturbed the plasma membrane lipid-ordered phase throughout the sarcolemma and was more pronounced in junctional membrane regions. Ouabain (1 µM), which specifically inhibits the Na-K-ATPase α2 isozyme in rodent skeletal muscles, produced similar lipid raft changes in control muscles but was ineffective in suspended muscles, which showed an initial loss of α2 Na-K-ATPase activity. Lipid rafts were able to recover with cholesterol supplementation, suggesting that disturbance results from cholesterol loss. Repetitive nerve stimulation also restores lipid rafts, specifically in the junctional sarcolemma region. Disuse locally lowered the resting intracellular Ca2+ concentration only near the neuromuscular junction of muscle fibers. Our results provide evidence to suggest that the ordering of lipid rafts strongly depends on motor nerve input and may involve interactions with the α2 Na-K-ATPase. Lipid raft disturbance, accompanied by intracellular Ca2+ dysregulation, is among the earliest remodeling events induced by skeletal muscle disuse.
Collapse
Affiliation(s)
- Alexey M Petrov
- Department of Normal Physiology, Kazan State Medical University, Kazan, Russia
| | - Violetta V Kravtsova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | | | - Alexander N Vasiliev
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Andrey L Zefirov
- Department of Normal Physiology, Kazan State Medical University, Kazan, Russia
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden; and
| | - Judith A Heiny
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Igor I Krivoi
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia;
| |
Collapse
|
89
|
Towards human exploration of space: the THESEUS review series on muscle and bone research priorities. NPJ Microgravity 2017. [PMID: 28649630 PMCID: PMC5445590 DOI: 10.1038/s41526-017-0013-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Without effective countermeasures, the musculoskeletal system is altered by the microgravity environment of long-duration spaceflight, resulting in atrophy of bone and muscle tissue, as well as in deficits in the function of cartilage, tendons, and vertebral disks. While inflight countermeasures implemented on the International Space Station have evidenced reduction of bone and muscle loss on low-Earth orbit missions of several months in length, important knowledge gaps must be addressed in order to develop effective strategies for managing human musculoskeletal health on exploration class missions well beyond Earth orbit. Analog environments, such as bed rest and/or isolation environments, may be employed in conjunction with large sample sizes to understand sex differences in countermeasure effectiveness, as well as interaction of exercise with pharmacologic, nutritional, immune system, sleep and psychological countermeasures. Studies of musculoskeletal biomechanics, involving both human subject and computer simulation studies, are essential to developing strategies to avoid bone fractures or other injuries to connective tissue during exercise and extravehicular activities. Animal models may be employed to understand effects of the space environment that cannot be modeled using human analog studies. These include studies of radiation effects on bone and muscle, unraveling the effects of genetics on bone and muscle loss, and characterizing the process of fracture healing in the mechanically unloaded and immuno-compromised spaceflight environment. In addition to setting the stage for evidence-based management of musculoskeletal health in long-duration space missions, the body of knowledge acquired in the process of addressing this array of scientific problems will lend insight into the understanding of terrestrial health conditions such as age-related osteoporosis and sarcopenia.
Collapse
|
90
|
Hu NF, Chang H, Du B, Zhang QW, Arfat Y, Dang K, Gao YF. Tetramethylpyrazine ameliorated disuse-induced gastrocnemius muscle atrophy in hindlimb unloading rats through suppression of Ca2+/ROS-mediated apoptosis. Appl Physiol Nutr Metab 2017; 42:117-127. [DOI: 10.1139/apnm-2016-0363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to examine the possible mechanism underlying the protective effect of tetramethylpyrazine (TMP) against disuse-induced muscle atrophy. Sprague−Dawley rats were randomly assigned to receive 14 days of hindlimb unloading (HLU, a model of disuse atrophy) or cage controls. The rats were given TMP (60 mg/kg body mass) or vehicle (water) by gavage. Compared with vehicle treatment, TMP significantly attenuated the loss of gastrocnemius muscle mass (−33.56%, P < 0.01), the decrease of cross-sectional area of slow fiber (−10.99%, P < 0.05) and fast fiber (−15.78%, P < 0.01) during HLU. Although TMP failed to further improve recovery of muscle function or fatigability compared with vehicle treatment, it can suppress the higher level of lactate (−22.71%, P < 0.01) induced by HLU. Besides, TMP could effectually reduce the increased protein expression of muscle RING-finger protein 1 induced by HLU (−14.52%, P < 0.01). Furthermore, TMP can ameliorate the calcium overload (−54.39%, P < 0.05), the increase of malondialdehyde content (−19.82%, P < 0.05), the decrease of superoxide dismutase activity (21.34%, P < 0.05), and myonuclear apoptosis (−78.22%, P < 0.01) induced by HLU. Moreover, TMP significantly reduced HLU-induced increase of Bax to B-cell lymphoma 2 (−36.36%, P < 0.01) and cytochrome c release (−36.16%, P < 0.05). In conclusion, TMP attenuated HLU-induced gastrocnemius muscle atrophy through suppression of Ca2+/reactive oxygen species increase and consequent proteolysis and apoptosis. Therefore, TMP might exhibit therapeutic effect against oxidative stress, cytosolic calcium overload, and mitochondrial damage in disuse-induced muscle atrophy.
Collapse
Affiliation(s)
- Nai-Fei Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| | - Bei Du
- Shaanxi Institute of International Trade and Commerce, Xian Yang 712046, China
| | - Quan-Wang Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| | - Yasir Arfat
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| | - Kai Dang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| | - Yun-Fang Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education, Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
| |
Collapse
|
91
|
The effect of long-term hindlimb unloading on the expression of risk neurogenes encoding elements of serotonin-, dopaminergic systems and apoptosis; comparison with the effect of actual spaceflight on mouse brain. Neurosci Lett 2017; 640:88-92. [DOI: 10.1016/j.neulet.2017.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/04/2017] [Accepted: 01/10/2017] [Indexed: 11/19/2022]
|
92
|
Xu Z, Sun W, Li Y, Ling S, Zhao C, Zhong G, Zhao D, Song J, Song H, Li J, You L, Nie G, Chang Y, Li Y. The regulation of iron metabolism by hepcidin contributes to unloading-induced bone loss. Bone 2017; 94:152-161. [PMID: 27686598 DOI: 10.1016/j.bone.2016.09.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/02/2016] [Accepted: 09/25/2016] [Indexed: 12/20/2022]
Abstract
Iron overload inhibits osteoblast function and promotes osteoclastogenesis. Hepcidin plays an important role in this process. The changes in iron content and the regulation of hepcidin under unloading-induced bone loss remain unknown. A hindlimb suspension model was adopted to simulate unloading-induced bone loss in mice. The results showed that iron deposition in both liver and bone was markedly increased in hindlimb unloaded mice, and was accompanied by the upregulation of osteoclast activity and downregulation of osteoblast activity. The iron chelator deferoxamine mesylate (DFO) reduced the iron content in bone and alleviated unloading-induced bone loss. The increased iron content in bone was mainly a result of the upregulation of transferrin receptor 1 (TfR1) and divalent metal transporter 1 with iron response element (DMT1+IRE), rather than changes in the iron transporter ferroportin 1 (FPN1). The hepcidin level in the liver was significantly higher, while the FPN1 level in the duodenum was substantially reduced. However, there were no changes in the FPN1 level in bone tissue. During hindlimb unloading, downregulation of hepcidin by siRNA increased iron uptake in bone and liver, which aggravated unloading-induced bone loss. In summary, these data show that unloading-induced bone loss was orchestrated by iron overload and coupled with the regulation of hepcidin by the liver.
Collapse
Affiliation(s)
- Zi Xu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Weijia Sun
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Chenyang Zhao
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Jinping Song
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Hailin Song
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Jinqiao Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China
| | - Linhao You
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China
| | - Guangjun Nie
- Key Laboratory of Chinese Academy of Sciences for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yanzhong Chang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China.
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, China.
| |
Collapse
|
93
|
Mirzoev TM, Tyganov SA, Petrova IO, Shenkman BS. The realization of a mechanical signal during gravitational unloading: The response of mTORC1 targets to eccentric contractions. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s0006350916050213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
94
|
Wang J, Wang F, Zhang P, Liu H, He J, Zhang C, Fan M, Chen X. PGC-1α over-expression suppresses the skeletal muscle atrophy and myofiber-type composition during hindlimb unloading. Biosci Biotechnol Biochem 2016; 81:500-513. [PMID: 27869526 DOI: 10.1080/09168451.2016.1254531] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Disuse leads to severe muscle atrophy and a slow-to-fast myofiber-type transition. PGC-1α (Peroxisome proliferator-activated receptor γ coactivator 1α) is documented to play an important role in muscle atrophy and slow-twitch myofiber determination. Transcription of atrophy-related Atrogin-1 by FoxO3 can be reduced by PGC-1α. While Smad3 augments FoxO3-induced Atrogin-1 and MuRF1 promoter activity. So PGC-1α, as a transcription co-activator, may regulate hindlimb unloading (HU)-induced myofiber-type transition and muscle atrophy through Smad3. Our results showed that transgenic PGC-1α mice resisted HU-induced muscle loss, atrophy-related genes expression, and slow-to-fast myofiber-type transition. Furthermore, over-expression of PGC-1α resisted the increase in pSmad3 during muscle atrophy in vivo and in vitro. And, PGC-1α over-expression inhibited the expression of atrogenes via suppressing the phosphorylation of Smad3 in vitro. Thus, PGC-1α is effective in regulating myofiber-type transition during HU, and it alleviates skeletal muscle atrophy partially through suppressing the activation of Smad3.
Collapse
Affiliation(s)
- Jing Wang
- a Department of Neurobiology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Fei Wang
- b National Key Laboratory of Human Factors Engineering , China Astronaut Research and Training Center , Beijing , China
| | - Peng Zhang
- c State Key Laboratory of Space Medicine Fundamentals and Application , China Astronaut Research and Training Center , Beijing , China
| | - Hongju Liu
- c State Key Laboratory of Space Medicine Fundamentals and Application , China Astronaut Research and Training Center , Beijing , China
| | - Jian He
- c State Key Laboratory of Space Medicine Fundamentals and Application , China Astronaut Research and Training Center , Beijing , China
| | - Chenyu Zhang
- d State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences, Nanjing University , Nanjing , China
| | - Ming Fan
- a Department of Neurobiology , School of Basic Medical Sciences, Capital Medical University , Beijing , China
| | - Xiaoping Chen
- b National Key Laboratory of Human Factors Engineering , China Astronaut Research and Training Center , Beijing , China.,c State Key Laboratory of Space Medicine Fundamentals and Application , China Astronaut Research and Training Center , Beijing , China
| |
Collapse
|
95
|
Strickland JC, Smith MA. Animal models of resistance exercise and their application to neuroscience research. J Neurosci Methods 2016; 273:191-200. [PMID: 27498037 PMCID: PMC5075509 DOI: 10.1016/j.jneumeth.2016.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/24/2016] [Accepted: 08/03/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Numerous studies have demonstrated that participation in regular resistance exercise (e.g., strength training) is associated with improvements in mental health, memory, and cognition. However, less is known about the neurobiological mechanisms mediating these effects. The goal of this mini-review is to describe and evaluate the available animal models of resistance exercise that may prove useful for examining CNS activity. NEW METHOD Various models have been developed to examine resistance exercise in laboratory animals. COMPARISON WITH EXISTING METHODS Resistance exercise models vary in how the resistance manipulation is applied, either through direct stimulation of the muscle (e.g., in situ models) or through behavior maintained by operant contingencies (e.g., whole organism models). Each model presents distinct advantages and disadvantages for examining central nervous system (CNS) activity, and consideration of these attributes is essential for the future investigation of underlying neurobiological substrates. RESULTS Potential neurobiological mechanisms mediating the effects of resistance exercise on pain, anxiety, memory, and drug use have been efficiently and effectively investigated using resistance exercise models that minimize stress and maximize the relative contribution of resistance over aerobic factors. CONCLUSIONS Whole organism resistance exercise models that (1) limit the use of potentially stressful stimuli and (2) minimize the contribution of aerobic factors will be critical for examining resistance exercise and CNS function.
Collapse
Affiliation(s)
| | - Mark A Smith
- Department of Psychology, Davidson College, Davidson, NC, USA; Program in Neuroscience, Davidson College, Davidson, NC, USA.
| |
Collapse
|
96
|
Pierre N, Appriou Z, Gratas-Delamarche A, Derbré F. From physical inactivity to immobilization: Dissecting the role of oxidative stress in skeletal muscle insulin resistance and atrophy. Free Radic Biol Med 2016; 98:197-207. [PMID: 26744239 DOI: 10.1016/j.freeradbiomed.2015.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 12/16/2022]
Abstract
In the literature, the terms physical inactivity and immobilization are largely used as synonyms. The present review emphasizes the need to establish a clear distinction between these two situations. Physical inactivity is a behavior characterized by a lack of physical activity, whereas immobilization is a deprivation of movement for medical purpose. In agreement with these definitions, appropriate models exist to study either physical inactivity or immobilization, leading thereby to distinct conclusions. In this review, we examine the involvement of oxidative stress in skeletal muscle insulin resistance and atrophy induced by, respectively, physical inactivity and immobilization. A large body of evidence demonstrates that immobilization-induced atrophy depends on the chronic overproduction of reactive oxygen and nitrogen species (RONS). On the other hand, the involvement of RONS in physical inactivity-induced insulin resistance has not been investigated. This observation outlines the need to elucidate the mechanism by which physical inactivity promotes insulin resistance.
Collapse
Affiliation(s)
- Nicolas Pierre
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Zephyra Appriou
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Arlette Gratas-Delamarche
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France
| | - Frédéric Derbré
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, Rennes 2 University - ENS Rennes, Bruz, France.
| |
Collapse
|
97
|
|
98
|
Macias BR, Lima F, Swift JM, Shirazi-Fard Y, Greene ES, Allen MR, Fluckey J, Hogan HA, Braby L, Wang S, Bloomfield SA. Simulating the Lunar Environment: Partial Weightbearing and High-LET Radiation-Induce Bone Loss and Increase Sclerostin-Positive Osteocytes. Radiat Res 2016; 186:254-63. [PMID: 27538114 DOI: 10.1667/rr13579.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Exploration missions to the Moon or Mars will expose astronauts to galactic cosmic radiation and low gravitational fields. Exposure to reduced weightbearing and radiation independently result in bone loss. However, no data exist regarding the skeletal consequences of combining low-dose, high-linear energy transfer (LET) radiation and partial weightbearing. We hypothesized that simulated galactic cosmic radiation would exacerbate bone loss in animals held at one-sixth body weight (G/6) without radiation exposure. Female BALB/cByJ four-month-old mice were randomly assigned to one of the following treatment groups: 1 gravity (1G) control; 1G with radiation; G/6 control; and G/6 with radiation. Mice were exposed to either silicon-28 or X-ray radiation. (28)Si radiation (300 MeV/nucleon) was administered at acute doses of 0 (sham), 0.17 and 0.5 Gy, or in three fractionated doses of 0.17 Gy each over seven days. X radiation (250 kV) was administered at acute doses of 0 (sham), 0.17, 0.5 and 1 Gy, or in three fractionated doses of 0.33 Gy each over 14 days. Bones were harvested 21 days after the first exposure. Acute 1 Gy X-ray irradiation during G/6, and acute or fractionated 0.5 Gy (28)Si irradiation during 1G resulted in significantly lower cancellous mass [percentage bone volume/total volume (%BV/TV), by microcomputed tomography]. In addition, G/6 significantly reduced %BV/TV compared to 1G controls. When acute X-ray irradiation was combined with G/6, distal femur %BV/TV was significantly lower compared to G/6 control. Fractionated X-ray irradiation during G/6 protected against radiation-induced losses in %BV/TV and trabecular number, while fractionated (28)Si irradiation during 1G exacerbated the effects compared to single-dose exposure. Impaired bone formation capacity, measured by percentage mineralizing surface, can partially explain the lower cortical bone thickness. Moreover, both partial weightbearing and (28)Si-ion exposure contribute to a higher proportion of sclerostin-positive osteocytes in cortical bone. Taken together, these data suggest that partial weightbearing and low-dose, high-LET radiation negatively impact maintenance of bone mass by lowering bone formation and increasing bone resorption. The impaired bone formation response is associated with sclerostin-induced suppression of Wnt signaling. Therefore, exposure to low-dose, high-LET radiation during long-duration spaceflight missions may reduce bone formation capacity, decrease cancellous bone mass and increase bone resorption. Future countermeasure strategies should aim to restore mechanical loads on bone to those experienced in one gravity. Moreover, low-doses of high-LET radiation during long-duration spaceflight should be limited or countermeasure strategies employed to mitigate bone loss.
Collapse
Affiliation(s)
| | | | | | | | | | - M R Allen
- g Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | - H A Hogan
- b Biomedical Engineering.,c Mechanical Engineering
| | | | - Suojin Wang
- f Statistics, Texas A&M University, College Station, Texas, 77843 and
| | - S A Bloomfield
- a Health and Kinesiology.,d Intercollegiate Faculty of Nutrition
| |
Collapse
|
99
|
Mirzoev TM, Tyganov SA, Shenkman BS. Akt-dependent and Akt-independent pathways are involved in protein synthesis activation during reloading of disused soleus muscle. Muscle Nerve 2016; 55:393-399. [DOI: 10.1002/mus.25235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/10/2016] [Accepted: 06/30/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Timur M. Mirzoev
- Myology Laboratory, Institute of Bio-Medical Problems of the Russian Academy of Sciences; 123007 Moscow Russian Federation
| | - Sergey A. Tyganov
- Myology Laboratory, Institute of Bio-Medical Problems of the Russian Academy of Sciences; 123007 Moscow Russian Federation
| | - Boris S. Shenkman
- Myology Laboratory, Institute of Bio-Medical Problems of the Russian Academy of Sciences; 123007 Moscow Russian Federation
| |
Collapse
|
100
|
Lippert T, Watson N, Ji X, Yasuhara T, Date I, Kaneko Y, Tajiri N, Borlongan CV. Detrimental effects of physical inactivity on neurogenesis. Brain Circ 2016; 2:80-85. [PMID: 30276277 PMCID: PMC6126252 DOI: 10.4103/2394-8108.186278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 02/25/2016] [Accepted: 03/04/2016] [Indexed: 01/01/2023] Open
Abstract
Patients diagnosed with neurological disorders exhibit a variety of physical and psychiatric symptoms, including muscle atrophy, general immobility, and depression. Patients who participate in physical rehabilitation at times show unexpected clinical improvement, which includes diminished depression and other stress-related behaviors. Regenerative medicine has advanced two major stem cell-based therapies for central nervous system (CNS) disorders, transplantation of exogenous stem cells, and enhancing the endogenous neurogenesis. The latter therapy utilizes a natural method of re-innervating the injured brain, which may mend neurological impairments. In this study, we examine how inactivity-induced atrophy, using the hindlimb suspension model, alters neurogenesis in rats. The hypothesis is that inactivity inhibits neurogenesis by decreasing circulation growth or trophic factors, such as vascular endothelial growth or neurotrophic factors. The restriction modifies neurogenesis and stem cell differentiation in the CNS, the stem cell microenvironment is examined by the trophic and growth factors, including stress-related proteins. Despite growing evidence revealing the benefits of “increased” exercise on neurogenesis, the opposing theory involving “physical inactivity,” which simulates pathological states, continues to be neglected. This novel theory will allow us to explore the effects on neurogenesis by an intransigent stem cell microenvironment likely generated by inactivity. 5-bromo-2-deoxyuridine labeling of proliferative cells, biochemical assays of serum, cerebrospinal fluid, and brain levels of trophic factors, growth factors, and stress-related proteins are suggested identifiers of neurogenesis, while evaluation of spontaneous movements will give insight into the psychomotor effects of inactivity. Investigations devised to show how in vivo stimulation, or lack thereof, affects the stem cell microenvironment are necessary to establish treatment methods to boost neurogenesis in bedridden patients.
Collapse
Affiliation(s)
- Trenton Lippert
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Nate Watson
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| |
Collapse
|