Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 18(10)2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-28994728

RESUMEN

Space radiation may pose a risk to skeletal health during subsequent aging. Irradiation acutely stimulates bone remodeling in mice, although the long-term influence of space radiation on bone-forming potential (osteoblastogenesis) and possible adaptive mechanisms are not well understood. We hypothesized that ionizing radiation impairs osteoblastogenesis in an ion-type specific manner, with low doses capable of modulating expression of redox-related genes. 16-weeks old, male, C57BL6/J mice were exposed to low linear-energy-transfer (LET) protons (150 MeV/n) or high-LET 56Fe ions (600 MeV/n) using either low (5 or 10 cGy) or high (50 or 200 cGy) doses at NASA's Space Radiation Lab. Five weeks or one year after irradiation, tissues were harvested and analyzed by microcomputed tomography for cancellous microarchitecture and cortical geometry. Marrow-derived, adherent cells were grown under osteoblastogenic culture conditions. Cell lysates were analyzed by RT-PCR during the proliferative or mineralizing phase of growth, and differentiation was analyzed by imaging mineralized nodules. As expected, a high dose (200 cGy), but not lower doses, of either 56Fe or protons caused a loss of cancellous bone volume/total volume. Marrow cells produced mineralized nodules ex vivo regardless of radiation type or dose; 56Fe (200 cGy) inhibited osteoblastogenesis by more than 90% (5 weeks and 1 year post-IR). After 5 weeks, irradiation (protons or 56Fe) caused few changes in gene expression levels during osteoblastogenesis, although a high dose 56Fe (200 cGy) increased Catalase and Gadd45. The addition of exogenous superoxide dismutase (SOD) protected marrow-derived osteoprogenitors from the damaging effects of exposure to low-LET (137Cs γ) when irradiated in vitro, but had limited protective effects on high-LET 56Fe-exposed cells. In sum, either protons or 56Fe at a relatively high dose (200 cGy) caused persistent bone loss, whereas only high-LET 56Fe increased redox-related gene expression, albeit to a limited extent, and inhibited osteoblastogenesis. Doses below 50 cGy did not elicit widespread responses in any parameter measured. We conclude that high-LET irradiation at 200 cGy impaired osteoblastogenesis and regulated steady-state gene expression of select redox-related genes during osteoblastogenesis, which may contribute to persistent bone loss.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Isótopos de Hierro/efectos adversos , Fenómenos Fisiológicos Musculoesqueléticos/efectos de la radiación , Osteogénesis/efectos de la radiación , Estrés Oxidativo , Exposición a la Radiación/efectos adversos , Animales , Relación Dosis-Respuesta en la Radiación , Expresión Génica/genética , Expresión Génica/efectos de la radiación , Transferencia Lineal de Energía , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis/genética , Oxidación-Reducción/efectos de la radiación , Protones/efectos adversos , Dosis de Radiación , Radiación Ionizante
2.
Sci Rep ; 14(1): 7334, 2024 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409284

RESUMEN

Exposure to cosmic ionizing radiation is an innate risk of the spaceflight environment that can cause DNA damage and altered cellular function. In astronauts, longitudinal monitoring of physiological systems and interactions between these systems are important to consider for mitigation strategies. In addition, assessments of sex-specific biological responses in the unique environment of spaceflight are vital to support future exploration missions that include both females and males. Here we assessed sex-specific, multi-system immune and endocrine responses to simulated cosmic radiation. For this, 24-week-old, male and female C57Bl/6J mice were exposed to simplified five-ion, space-relevant galactic cosmic ray (GCRsim) radiation at 15 and 50 cGy, to simulate predicted radiation exposures that would be experienced during lunar and Martian missions, respectively. Blood and adrenal tissues were collected at 3- and 14-days post-irradiation for analysis of immune and endocrine biosignatures and pathways. Sexually dimorphic adrenal gland weights and morphology, differential total RNA expression with corresponding gene ontology, and unique immune phenotypes were altered by GCRsim. In brief, this study offers new insights into sexually dimorphic immune and endocrine kinetics following simulated cosmic radiation exposure and highlights the necessity for personalized translational approaches for astronauts during exploration missions.


Asunto(s)
Radiación Cósmica , Marte , Vuelo Espacial , Ratones , Masculino , Femenino , Animales , Medio Ambiente Extraterrestre , Caracteres Sexuales , Radiación Ionizante , Astronautas , Radiación Cósmica/efectos adversos , Inmunidad
3.
Front Endocrinol (Lausanne) ; 14: 1141906, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37455901

RESUMEN

Introduction: Reductions in energy availability leading to weight loss can induce loss of bone and impact important endocrine regulators of bone integrity. We sought to elucidate whether endurance exercise (EX) can mitigate bone loss observed in sedentary (SED) skeletally mature rodents subjected to graded energy deficits. Methods: Female virgin rats (n=84, 5-mo-old; 12/group) were randomized to baseline controls and either sedentary (SED) or exercise (EX) conditions, and within each exercise status to adlib-fed (ADLIB), or moderate (MOD) or severe (SEV) energy restriction diets for 12 weeks. Rats assigned to EX groups performed treadmill running to increase weekly energy expenditure by 10%. MOD-ER-SED, SEV-ER-SED, MOD-ER-EX and SEV-ER-EX were fed modified AIN93M diets with 20%, 40% 10%, and 30% less energy content, respectively, with 100% of all other nutrients provided. Results: Energy availability (EA) was effectively reduced by ~14% and ~30% in the MOD-ER and SEV-ER groups, respectively. MOD-ER for 12 weeks resulted in few negative impacts on bone and, except for serum leptin in MOD-ER-SED rats, no significant changes in endocrine factors. By contrast, SEV-ER in SED rats resulted in significantly lower total body and femoral neck bone mass, and reduced serum estradiol, IGF-1 and leptin. EX rats experiencing the same reduction in energy availability as SEV-ER-SED exhibited higher total body mass, lean mass, total BMC, and higher serum IGF-1 at the end of 12 weeks. Bone mechanical properties at 3 bone sites (mid-femur, distal femur, femoral neck) were minimally impacted by ER but positively affected by EX. Discussion: These findings indicate that combining increased EX energy expenditure with smaller reductions in energy intake to achieve a targeted reduction in EA provides some protection against loss of bone mass and lean mass in skeletally mature female rats, likely due to better preservation of circulating IGF-1, and that bone mechanical integrity is not significantly degraded with either moderate or severe reduced EA.


Asunto(s)
Leptina , Condicionamiento Físico Animal , Animales , Femenino , Ratas , Huesos , Factor I del Crecimiento Similar a la Insulina , Condicionamiento Físico Animal/fisiología
4.
Life (Basel) ; 13(5)2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37240858

RESUMEN

Exposure to space galactic cosmic radiation is a principal consideration for deep space missions. While the effects of space irradiation on the nervous system are not fully known, studies in animal models have shown that exposure to ionizing radiation can cause neuronal damage and lead to downstream cognitive and behavioral deficits. Cognitive health implications put humans and missions at risk, and with the upcoming Artemis missions in which female crew will play a major role, advance critical analysis of the neurological and performance responses of male and female rodents to space radiation is vital. Here, we tested the hypothesis that simulated Galactic Cosmic Radiation (GCRSim) exposure disrupts species-typical behavior in mice, including burrowing, rearing, grooming, and nest-building that depend upon hippocampal and medial prefrontal cortex circuitry. Behavior comprises a remarkably well-integrated representation of the biology of the whole animal that informs overall neural and physiological status, revealing functional impairment. We conducted a systematic dose-response analysis of mature (6-month-old) male and female mice exposed to either 5, 15, or 50 cGy 5-ion GCRSim (H, Si, He, O, Fe) at the NASA Space Radiation Laboratory (NSRL). Behavioral performance was evaluated at 72 h (acute) and 91-days (delayed) postradiation exposure. Specifically, species-typical behavior patterns comprising burrowing, rearing, and grooming as well as nest building were analyzed. A Neuroscore test battery (spontaneous activity, proprioception, vibrissae touch, limb symmetry, lateral turning, forelimb outstretching, and climbing) was performed at the acute timepoint to investigate early sensorimotor deficits postirradiation exposure. Nest construction, a measure of neurological and organizational function in rodents, was evaluated using a five-stage Likert scale 'Deacon' score that ranged from 1 (a low score where the Nestlet is untouched) to 5 (a high score where the Nestlet is completely shredded and shaped into a nest). Differential acute responses were observed in females relative to males with respect to species-typical behavior following 15 cGy exposure while delayed responses were observed in female grooming following 50 cGy exposure. Significant sex differences were observed at both timepoints in nest building. No deficits in sensorimotor behavior were observed via the Neuroscore. This study revealed subtle, sexually dimorphic GCRSim exposure effects on mouse behavior. Our analysis provides a clearer understanding of GCR dose effects on species typical, sensorimotor and organizational behaviors at acute and delayed timeframes postirradiation, thereby setting the stage for the identification of underlying cellular and molecular events.

5.
NPJ Microgravity ; 9(1): 75, 2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37723136

RESUMEN

Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).

6.
Life Sci Space Res (Amst) ; 37: 39-49, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37087178

RESUMEN

BACKGROUND: The limitations to prolonged spaceflight include unloading-induced atrophy of the musculoskeletal system which may be enhanced by exposure to the space radiation environment. Previous results have concluded that partial gravity, comparable to the Lunar surface, may have detrimental effects on skeletal muscle. However, little is known if these outcomes are exacerbated by exposure to low-dose rate, high-energy radiation common to the space environment. Therefore, the present study sought to determine the impact of highly charge, high-energy (HZE) radiation on skeletal muscle when combined with partial weightbearing to simulate Lunar gravity. We hypothesized that partial unloading would compromise skeletal muscle and these effects would be exacerbated by radiation exposure. METHODS: For month old female BALB/cByJ mice were -assigned to one of 2 groups; either full weight bearing (Cage Controls, CC) or partial weight bearing equal to 1/6th bodyweight (G/6). Both groups were then divided to receive either a single whole body absorbed dose of 0.5 Gy of 300 MeV 28Si ions (RAD) or a sham treatment (SHAM). Radiation exposure experiments were performed at the NASA Space Radiation Laboratory (NSRL) located at Brookhaven National Laboratory on Day 0, followed by 21 d of CC or G/6 loading. Muscles of the hind limb were used to measure protein synthesis and other histological measures. RESULTS: Twenty-one days of Lunar gravity (G/6) resulted in lower soleus, plantaris, and gastrocnemius muscle mass. Radiation exposure did not further impact muscle mass. 28Si exposure in normal ambulatory animals (RAD+CC) did not impact gastrocnemius muscle mass when compared to SHAM+CC (p>0.05), but did affect the soleus, where mass was higher following radiation compared to SHAM (p<0.05). Mixed gastrocnemius muscle protein synthesis was lower in both unloading groups. Fiber type composition transitioned towards a faster isoform with partial unloading and was not further impacted by radiation. The combined effects of partial loading and radiation partially mitigated fiber cross-sectional area when compared to partial loading alone. Radiation and G/6 reduced the total number of myonuclei per fiber while leading to elevated BrdU content of skeletal muscle. Similarly, unloading and radiation resulted in higher collagen content of muscle when compared to controls, but the effects of combined exposure were not additive. CONCLUSIONS: The results of this study confirm that partial weightbearing causes muscle atrophy, in part due to reductions of muscle protein synthesis in the soleus and gastrocnemius as well as reduced peripheral nuclei per fiber. Additionally, we present novel data illustrating 28Si exposure reduced nuclei in muscle fibers despite higher satellite cell fusion, but did not exacerbate muscle atrophy, CSA changes, or collagen content. In conclusion, both partial loading and HZE radiation can negatively impact muscle morphology.


Asunto(s)
Iones Pesados , Ratones , Animales , Femenino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Atrofia Muscular/metabolismo , Colágeno/metabolismo , Colágeno/farmacología , Suspensión Trasera/efectos adversos , Suspensión Trasera/fisiología
7.
Cell Rep ; 42(5): 112299, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37080202

RESUMEN

Understanding the axis of the human microbiome and physiological homeostasis is an essential task in managing deep-space-travel-associated health risks. The NASA-led Rodent Research 5 mission enabled an ancillary investigation of the gut microbiome, varying exposure to microgravity (flight) relative to ground controls in the context of previously shown bone mineral density (BMD) loss that was observed in these flight groups. We demonstrate elevated abundance of Lactobacillus murinus and Dorea sp. during microgravity exposure relative to ground control through whole-genome sequencing and 16S rRNA analyses. Specific functionally assigned gene clusters of L. murinus and Dorea sp. capable of producing metabolites, lactic acid, leucine/isoleucine, and glutathione are enriched. These metabolites are elevated in the microgravity-exposed host serum as shown by liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. Along with BMD loss, ELISA reveals increases in osteocalcin and reductions in tartrate-resistant acid phosphatase 5b signifying additional loss of bone homeostasis in flight.


Asunto(s)
Microbioma Gastrointestinal , Vuelo Espacial , Humanos , ARN Ribosómico 16S/genética , Cromatografía Liquida , Viaje , Espectrometría de Masas en Tándem
8.
Sci Rep ; 10(1): 2336, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047211

RESUMEN

Animal models are useful for exploring the health consequences of prolonged spaceflight. Capabilities were developed to perform experiments in low earth orbit with on-board sample recovery, thereby avoiding complications caused by return to Earth. For NASA's Rodent Research-1 mission, female mice (ten 32 wk C57BL/6NTac; ten 16 wk C57BL/6J) were launched on an unmanned vehicle, then resided on the International Space Station for 21/22d or 37d in microgravity. Mice were euthanized on-orbit, livers and spleens dissected, and remaining tissues frozen in situ for later analyses. Mice appeared healthy by daily video health checks and body, adrenal, and spleen weights of 37d-flight (FLT) mice did not differ from ground controls housed in flight hardware (GC), while thymus weights were 35% greater in FLT than GC. Mice exposed to 37d of spaceflight displayed elevated liver mass (33%) and select enzyme activities compared to GC, whereas 21/22d-FLT mice did not. FLT mice appeared more physically active than respective GC while soleus muscle showed expected atrophy. RNA and enzyme activity levels in tissues recovered on-orbit were of acceptable quality. Thus, this system establishes a new capability for conducting long-duration experiments in space, enables sample recovery on-orbit, and avoids triggering standard indices of chronic stress.


Asunto(s)
Peso Corporal , Hígado/metabolismo , Vuelo Espacial , Ingravidez , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Factores de Tiempo
9.
Appl Physiol Nutr Metab ; 45(3): 264-274, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31340136

RESUMEN

Muscle disuse impairs muscle quality and is associated with increased mortality. Little is known regarding additive effects of multiple bouts of disuse, which is a common occurrence in patients experiencing multiple surgeries. Mitochondrial quality is vital to muscle health and quality; however, to date mitochondrial quality control has not been investigated following multiple bouts of disuse. Therefore, the purpose of this study was to investigate mitochondrial quality controllers during multiple bouts of disuse by hindlimb unloading. Male rats (n ∼ 8/group) were assigned to the following groups: hindlimb unloading for 28 days, hindlimb unloading with 56 days of reloading, 2 bouts of hindlimb unloading separated by a recovery phase of 56 days of reloading, 2 bouts of hindlimb unloading and recovery after each disuse, or control animals with no unloading. At designated time points, tissues were collected for messenger RNA and protein analysis of mitochondrial quality. Measures of mitochondrial biogenesis, such as proliferator-activated receptor gamma coactivator 1 alpha, decreased 30%-40% with unloading with no differences noted between unloading conditions. Measures of mitochondrial translation were 40%-50% lower in unloading conditions, with no differences noted between bouts of unloading. Measures of mitophagy were 40%-50% lower with reloading, with no differences noted between reloading conditions. In conclusion, disuse causes alterations in measures of mitochondrial quality; however, multiple bouts of disuse does not appear to have additive effects. Novelty Disuse atrophy causes multiple alterations to mitochondrial quality control. With sufficient recovery most detriments to mitochondrial quality control are fixed. In general, multiple bouts of disuse do not produce additive effects.


Asunto(s)
Suspensión Trasera/métodos , Mitocondrias Musculares/fisiología , Atrofia Muscular/fisiopatología , Biogénesis de Organelos , Animales , Modelos Animales de Enfermedad , Miembro Posterior/metabolismo , Miembro Posterior/fisiopatología , Suspensión Trasera/estadística & datos numéricos , Masculino , Músculo Esquelético/fisiopatología , Ratas , Ratas Sprague-Dawley
10.
Cell Rep ; 33(10): 108448, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33242410

RESUMEN

We have identified and validated a spaceflight-associated microRNA (miRNA) signature that is shared by rodents and humans in response to simulated, short-duration and long-duration spaceflight. Previous studies have identified miRNAs that regulate rodent responses to spaceflight in low-Earth orbit, and we have confirmed the expression of these proposed spaceflight-associated miRNAs in rodents reacting to simulated spaceflight conditions. Moreover, astronaut samples from the NASA Twins Study confirmed these expression signatures in miRNA sequencing, single-cell RNA sequencing (scRNA-seq), and single-cell assay for transposase accessible chromatin (scATAC-seq) data. Additionally, a subset of these miRNAs (miR-125, miR-16, and let-7a) was found to regulate vascular damage caused by simulated deep space radiation. To demonstrate the physiological relevance of key spaceflight-associated miRNAs, we utilized antagomirs to inhibit their expression and successfully rescue simulated deep-space-radiation-mediated damage in human 3D vascular constructs.


Asunto(s)
MicroARN Circulante/genética , MicroARNs/genética , Ingravidez/efectos adversos , Animales , Femenino , Expresión Génica , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Ratas , Análisis de Secuencia de ARN/métodos , Vuelo Espacial , Transcriptoma/genética , Simulación de Ingravidez/métodos
11.
J Vis Exp ; (143)2019 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-30688299

RESUMEN

Performing biological experiments in space requires special accommodations and procedures to ensure that these investigations are performed effectively and efficiently. Moreover, given the infrequency of these experiments it is imperative that their impacts be maximized. The rapid advancement of omics technologies offers an opportunity to dramatically increase the volume of data produced from precious spaceflight specimens. To capitalize on this, NASA has developed the GeneLab platform to provide unrestricted access to spaceflight omics data and encourage its widespread analysis. Rodents (both rats and mice) are common model organisms used by scientists to investigate space-related biological impacts. The enclosure that house rodents during spaceflight are called Rodent Habitats (formerly Animal Enclosure Modules), and are substantially different from standard vivarium cages in their dimensions, air flow, and access to water and food. In addition, due to environmental and atmospheric conditions on the International Space Station (ISS), animals are exposed to a higher CO2 concentration. We recently reported that mice in the Rodent Habitats experience large changes in their transcriptome irrespective of whether animals were on the ground or in space. Furthermore, these changes were consistent with a hypoxic response, potentially driven by higher CO2 concentrations. Here we describe how a typical rodent experiment is performed in space, how omics data from these experiments can be accessed through the GeneLab platform, and how to identify key factors in this data. Using this process, any individual can make critical discoveries that could change the design of future space missions and activities.


Asunto(s)
Vuelo Espacial , Transcriptoma , Ingravidez , Acceso a la Información , Animales , Ratones , Ratas , Estados Unidos , United States National Aeronautics and Space Administration
12.
J Appl Physiol (1985) ; 125(5): 1456-1467, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30091665

RESUMEN

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.


Asunto(s)
Suspensión Trasera/fisiología , Proteínas Musculares/biosíntesis , Músculo Esquelético/fisiología , Aminoácidos/metabolismo , Animales , Colágeno/metabolismo , Masculino , Músculo Esquelético/diagnóstico por imagen , Ratas Sprague-Dawley , Transducción de Señal
13.
Life Sci Space Res (Amst) ; 16: 52-62, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29475520

RESUMEN

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.


Asunto(s)
Huesos/fisiopatología , Fémur/fisiopatología , Curación de Fractura , Vuelo Espacial/instrumentación , Simulación del Espacio , Animales , Fenómenos Biomecánicos , Huesos/efectos de la radiación , Fémur/efectos de la radiación , Curación de Fractura/efectos de la radiación , Suspensión Trasera , Masculino , Ratones , Ratones Endogámicos C57BL , Ingravidez , Microtomografía por Rayos X
14.
Radiat Res ; 185(3): 257-66, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26930379

RESUMEN

Weightlessness during spaceflight leads to functional changes in resistance arteries and loss of cancellous bone, which may be potentiated by radiation exposure. The purpose of this study was to assess the effects of hindlimb unloading (HU) and total-body irradiation (TBI) on the vasomotor responses of skeletal muscle arteries. Male C57BL/6 mice were assigned to control, HU (13-16 days), TBI (1 Gy (56)Fe, 600 MeV, 10 cGy/min) and HU-TBI groups. Gastrocnemius muscle feed arteries were isolated for in vitro study. Endothelium-dependent (acetylcholine) and -independent (Dea-NONOate) vasodilator and vasoconstrictor (KCl, phenylephrine and myogenic) responses were evaluated. Arterial endothelial nitric oxide synthase (eNOS), superoxide dismutase-1 (SOD-1) and xanthine oxidase (XO) protein content and tibial cancellous bone microarchitecture were quantified. Endothelium-dependent and -independent vasodilator responses were impaired in all groups relative to control, and acetylcholine-induced vasodilation was lower in the HU-TBI group relative to that in the HU and TBI groups. Reductions in endothelium-dependent vasodilation correlated with a lower cancellous bone volume fraction. Nitric oxide synthase inhibition abolished all group differences in endothelium-dependent vasodilation. HU and HU-TBI resulted in decreases in eNOS protein levels, while TBI and HU-TBI produced lower SOD-1 and higher XO protein content. Vasoconstrictor responses were not altered. Reductions in NO bioavailability (eNOS), lower anti-oxidant capacity (SOD-1) and higher pro-oxidant capacity (XO) may contribute to the deficits in NOS signaling in skeletal muscle resistance arteries. These findings suggest that the combination of insults experienced in spaceflight leads to impairment of vasodilator function in resistance arteries that is mediated through deficits in NOS signaling.


Asunto(s)
Músculo Esquelético/efectos de la radiación , Exposición a la Radiación , Vasodilatación/efectos de la radiación , Sistema Vasomotor/efectos de la radiación , Animales , Arterias/metabolismo , Arterias/efectos de la radiación , Miembro Posterior/metabolismo , Miembro Posterior/efectos de la radiación , Humanos , Masculino , Ratones , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Vuelo Espacial , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Vasodilatadores/administración & dosificación , Sistema Vasomotor/metabolismo , Irradiación Corporal Total , Xantina Oxidasa/metabolismo
15.
Bone ; 81: 260-269, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26191778

RESUMEN

During spaceflight, astronauts will be exposed to a complex mixture of ionizing radiation that poses a risk to their health. Exposure of rodents to ionizing radiation on Earth causes bone loss and increases osteoclasts in cancellous tissue, but also may cause persistent damage to stem cells and osteoprogenitors. We hypothesized that ionizing radiation damages skeletal tissue despite a prolonged recovery period, and depletes the ability of cells in the osteoblast lineage to respond at a later time. The goal of the current study was to test if irradiation prevents bone accrual and bone formation induced by an anabolic mechanical stimulus. Tibial axial compression was used as an anabolic stimulus after irradiation with heavy ions. Mice (male, C57BL/6J, 16 weeks) were exposed to high atomic number, high energy (HZE) iron ions ((56)Fe, 2 Gy, 600 MeV/ion) (IR, n=5) or sham-irradiated (Sham, n=5). In vivo axial loading was initiated 5 months post-irradiation; right tibiae in anesthetized mice were subjected to an established protocol known to stimulate bone formation (cyclic 9N compressive pulse, 60 cycles/day, 3 day/wk for 4 weeks). In vivo data showed no difference due to irradiation in the apparent stiffness of the lower limb at the initiation of the axial loading regimen. Axial loading increased cancellous bone volume by microcomputed tomography and bone formation rate by histomorphometry in both sham and irradiated animals, with a main effect of axial loading determined by two-factor ANOVA with repeated measure. There were no effects of radiation in cancellous bone microarchitecture and indices of bone formation. At the tibia diaphysis, results also revealed a main effect of axial loading on structure. Furthermore, irradiation prevented axial loading-induced stimulation of bone formation rate at the periosteal surface of cortical tissue. In summary, axial loading stimulated the net accrual of cancellous and cortical mass and increased cancellous bone formation rate despite prior exposure to ionizing radiation, in this case, HZE particles. Our findings suggest that mechanical stimuli may prove an effective treatment to improve skeletal structure following exposure to ionizing radiation.


Asunto(s)
Metabolismo/fisiología , Metabolismo/efectos de la radiación , Osteogénesis/fisiología , Osteogénesis/efectos de la radiación , Radiación Ionizante , Soporte de Peso/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Tibia/metabolismo , Tibia/efectos de la radiación
16.
J Interferon Cytokine Res ; 35(6): 480-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25734366

RESUMEN

Exposure to ionizing radiation can cause rapid mineral loss and increase bone-resorbing osteoclasts within metabolically active, cancellous bone tissue leading to structural deficits. To better understand mechanisms involved in rapid, radiation-induced bone loss, we determined the influence of total body irradiation on expression of select cytokines known both to stimulate osteoclastogenesis and contribute to inflammatory bone disease. Adult (16 week), male C57BL/6J mice were exposed to either 2 Gy gamma rays ((137)Cs, 0.8 Gy/min) or heavy ions ((56)Fe, 600MeV, 0.50-1.1 Gy/min); this dose corresponds to either a single fraction of radiotherapy (typical total dose is ≥10 Gy) or accumulates over long-duration interplanetary missions. Serum, marrow, and mineralized tissue were harvested 4 h-7 days later. Gamma irradiation caused a prompt (2.6-fold within 4 h) and persistent (peaking at 4.1-fold within 1 day) rise in the expression of the obligate osteoclastogenic cytokine, receptor activator of nuclear factor kappa-B ligand (Rankl), within marrow cells over controls. Similarly, Rankl expression peaked in marrow cells within 3 days of iron exposure (9.2-fold). Changes in Rankl expression induced by gamma irradiation preceded and overlapped with a rise in expression of other pro-osteoclastic cytokines in marrow (eg, monocyte chemotactic protein-1 increased by 11.9-fold, and tumor necrosis factor-alpha increased by 1.7-fold over controls). The ratio, Rankl/Opg, in marrow increased by 1.8-fold, a net pro-resorption balance. In the marrow, expression of the antioxidant transcription factor, Nfe2l2, strongly correlated with expression levels of Nfatc1, Csf1, Tnf, and Rankl. Radiation exposure increased a serum marker of bone resorption (tartrate-resistant acid phosphatase) and led to cancellous bone loss (16% decrement after 1 week). We conclude that total body irradiation (gamma or heavy-ion) caused temporal elevations in the concentrations of specific genes expressed within marrow and mineralized tissue related to bone resorption, including select cytokines that lead to osteoclastogenesis and elevated resorption; this is likely to account for rapid and progressive deterioration of cancellous microarchitecture following exposure to ionizing radiation.


Asunto(s)
Médula Ósea/efectos de la radiación , Resorción Ósea/genética , Huesos/efectos de la radiación , Rayos gamma/efectos adversos , Osteoclastos/efectos de la radiación , Fosfatasa Ácida/genética , Fosfatasa Ácida/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/metabolismo , Huesos/patología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Regulación de la Expresión Génica , Isoenzimas/genética , Isoenzimas/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Transducción de Señal , Fosfatasa Ácida Tartratorresistente , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Irradiación Corporal Total
17.
Bone ; 66: 296-305, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24929241

RESUMEN

Spaceflight provides a unique environment for skeletal tissue causing decrements in structural and densitometric properties of bone. Previously, we used the adult hindlimb unloaded (HU) rat model to show that previous exposure to HU had minimal effects on bone structure after a second HU exposure followed by recovery. Furthermore, we found that the decrements during second HU exposure were milder than the initial HU cycle. In this study, we used a moderate intensity resistance exercise protocol as an anabolic stimulus during recovery to test the hypothesis that resistance exercise following an exposure to HU will significantly enhance recovery of densitometric, structural, and, more importantly, mechanical properties of trabecular and cortical bone. We also hypothesized that resistance exercise during recovery, and prior to the second unloading period, will mitigate the losses during the second exposure. The hypothesis that exercise during recovery following hindlimb unloading will improve bone quality was supported by our data, as total BMC, total vBMD, and cancellous bone formation at the proximal tibia metaphysis increased significantly during exercise period, and total BMC/vBMD exceeded age-matched control and non-exercised values significantly by the end of recovery. However, our results did not support the hypothesis that resistance exercise prior to a subsequent unloading period will mitigate the detrimental effects of the second exposure, as the losses during the second exposure in total BMC, total vBMD, and cortical area at the proximal tibia metaphysis for the exercised animals were similar to those of the non-exercised group. Therefore, exercise did not mitigate effects of the second HU exposure in terms of pre-to-post HU changes in these variables, but it did produce beneficial effects in a broader sense.


Asunto(s)
Trastornos Musculares Atróficos/fisiopatología , Condicionamiento Físico Animal , Tibia/fisiopatología , Animales , Biomarcadores/metabolismo , Fenómenos Biomecánicos , Peso Corporal , Resorción Ósea/sangre , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Resorción Ósea/fisiopatología , Densitometría , Masculino , Trastornos Musculares Atróficos/sangre , Trastornos Musculares Atróficos/diagnóstico por imagen , Trastornos Musculares Atróficos/patología , Ratas Sprague-Dawley , Tibia/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Microtomografía por Rayos X
18.
Bone ; 52(1): 433-43, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23017660

RESUMEN

Profound bone loss at weight bearing sites is a primary effect of long-duration spaceflight. Moreover, a significant increase in estimated fracture risk remains even 1 year after returning to Earth; hence, it is important to define how quickly bone integrity can recover following prolonged disuse. This study characterized the loss and recovery dynamics of bone following a period of rodent hindlimb unloading in three anatomic sites. We hypothesized that the rat femoral neck would exhibit a discordant recovery dynamic most similar to that observed in astronauts' proximal femur; that is, bone mineral content (absolute mass) at this site would recover faster and more completely than would bone density and cortical area, and they will all recover before bone strength does. We characterized loss and long-term recovery of densitometric properties at the femoral neck, proximal tibia metaphysis, and tibia diaphysis, and also mechanical properties at the femoral neck and tibia diaphysis for which mechanical testing is amenable. We assessed the relationship between calculated strength indices and measured mechanical properties. Adult male Sprague-Dawley rats (6 months) were assigned to baseline, age-matched control (AC), and hindlimb unloaded (HU) groups. The HU group was unloaded for 28 days and then returned to normal cage activity for 84 days of weight bearing recovery (3 times the duration of HU). Fifteen animals were euthanized from each of the HU and AC groups on days 28, 56, 84, and 112 of the study. At baseline and then every 28 days in vivo longitudinal pQCT scans were taken at proximal tibia metaphysis (PTM) and tibia diaphysis (TD); ex vivo pQCT scans were taken later at the femoral neck (FN). TD and FN were tested to failure to measure mechanical properties. The hypothesis that the femoral neck in rats will exhibit a discordant recovery dynamic most similar to that observed in astronauts' proximal femurs was not supported by our data. At the femoral neck, densitometric and geometric variables (total BMC, total vBMD, cancellous vBMD, and cortical area) recovered to age-matched control levels after a recovery period twice the duration of unloading. Contrary to our hypothesis, changes in densitometric variables at the PTM provided a better model for changes in the human femoral neck with prolonged weightlessness. Following 28 days of HU, PTM total BMC recovered to age-matched control levels after roughly two times the duration of unloading; however, total vBMD did not recover even after three recovery periods. Cortical thinning occurred at the PTM following HU likely due to inhibition of periosteal growth; cortical shell thickness did not recover even after three recovery periods. Calculated strength indices suggested a loss in strength at the tibial diaphysis, which was not confirmed with direct testing of mechanical properties. HU had no effect on maximum fracture force at mid-tibia diaphysis; however, femoral neck experienced a significant loss of maximum force due to unloading that fully recovered after 28 days. Estimated strength indices for the femoral neck suggested a recovery period of 56 days in contrast to the 28-day recovery that was observed with mechanical testing. However, the inaccuracy of strength indices vs. directly measured mechanical properties highlights the continued importance of ground based animal models and mechanical testing. Our results demonstrate that the PTM in the rat better matches loss and recovery dynamics observed in astronauts' proximal femur than does the rat FN, at least in terms of densitometric variables. More complete utility of the rat PTM as a model in this case, however, depends upon meaningful characterization of changes in mechanical properties as well.


Asunto(s)
Densidad Ósea , Huesos/fisiopatología , Animales , Fenómenos Biomecánicos , Masculino , Ratas , Ratas Sprague-Dawley , Tomografía Computarizada por Rayos X/métodos
19.
Bone ; 56(2): 461-73, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23871849

RESUMEN

Extended periods of inactivity cause severe bone loss and concomitant deterioration of the musculoskeletal system. Considerable research has been aimed at better understanding the mechanisms and consequences of bone loss due to unloading and the associated effects on strength and fracture risk. One factor that has not been studied extensively but is of great interest, particularly for human spaceflight, is how multiple or repeated exposures to unloading and reloading affect the skeleton. Space agencies worldwide anticipate increased usage of repeat-flier crewmembers, and major thrust of research has focused on better understanding of microgravity effects on loss of bone density at weightbearing skeletal sites; however there is limited data available on repeat microgravity exposure. The adult hindlimb unloaded (HU) rat model was used to determine how an initial unloading cycle will affect a subsequent exposure to disuse and recovery thereafter. Animals underwent 28 days of HU starting at 6 months of age followed by 56 days of recovery, and then another 28 days of HU with 56 days of recovery. In vivo longitudinal pQCT was used to quantify bone morphological changes, and ex vivo µCT was used to quantify trabecular microarchitecture and cortical shell geometry at the proximal tibia metaphysis (PTM). The mechanical properties of trabecular bone were examined by the reduced platen compression mechanical test. The hypothesis that the initial HU exposure will mitigate decrements in bone mass and density for the second HU exposure was supported as pre- to post-HU declines in total BMC, total vBMD, and cortical area by in vivo pQCT at the proximal tibia metaphysis were milder for the second HU (and not significant) compared to an age-matched single HU (3% vs. 6%, 2% vs. 6%, and 2% vs. 6%, respectively). In contrast, the hypothesis was not supported at the microarchitectural level as losses in BV/TV and Tb.Th. were similar during 2nd HU exposure and age-matched single HU. Recovery with respect to post-HU values and compared to aging controls for total BMC, vBMD and cortical area were slower in older animals exposed to single or double HU cycles compared to recovery of younger animals exposed to a single HU bout. Despite milder recovery at the older age, there was no difference between unloaded animals and controls at the end of second recovery period. Therefore, the data did not support the hypothesis that two cycles of HU exposure with recovery would have a net negative effect. Mechanical properties of trabecular bone were affected more severely than densitometric measures (35% loss in trabecular bone ultimate stress vs. 9% loss in trabecular vBMD), which can be attributed most prominently to reductions in trabecular bone density and tissue mineral density. Together, our data demonstrate that initial exposure to mechanical unloading does not exacerbate bone loss during a subsequent unloading period and two cycles of unloading followed by recovery do not have a cumulative net negative effect on total bone mineral content and density as measured by pQCT at the proximal tibia metaphysis.


Asunto(s)
Suspensión Trasera/fisiología , Tibia/fisiología , Animales , Densidad Ósea/fisiología , Masculino , Ratones Endogámicos C57BL , Ratas , Simulación de Ingravidez
20.
Med Sci Sports Exerc ; 45(11): 2052-60, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23657172

RESUMEN

PURPOSE: The purpose of this study was to investigate whether partial weight-bearing activity, at either one-sixth or one-third of body mass, blunts the deleterious effects of simulated microgravity (0G) after 21 d on muscle mass and quantitative/qualitative measures of bone. METHODS: Using a novel, previously validated partial weight-bearing suspension device, mice were subjected to 16% (G/3, i.e., simulated lunar gravity) or 33% (G/6, i.e., simulated Martian gravity) weight bearing for 21 d. One gravity control (1G, i.e., Earth gravity) and tail-suspended mice (0G, i.e., simulated microgravity) served as controls to compare the effects of simulated lunar and Martian gravity to both Earth and microgravity. RESULTS: Simulated microgravity (0G) resulted in an 8% reduction in body mass and a 28% lower total plantarflexor muscle mass (for both, P < 0.01) as compared with 1G controls, but one-sixth and one-third partial weight-bearing activity attenuated losses. Relative to 1G controls, trabecular bone volume fraction (-9% to -13%) and trabecular thickness (-10% to -14%) were significantly lower in all groups (P < 0.01). In addition, cancellous and cortical bone formation rates (BFR) were lower in all reduced weight-bearing groups compared with 1G controls (-46% to -57%, trabecular BFR; -73% to -85%, cortical BFR; P < 0.001). Animals experiencing one-third but not one-sixth weight bearing exhibited attenuated deficits in femoral neck mechanical strength associated with 0G. CONCLUSION: These results suggest that partial weight bearing (up to 33% of body mass) is not sufficient to protect against bone loss observed with simulated 0 g but does mitigate reductions in soleus mass in skeletally mature female mice.


Asunto(s)
Resorción Ósea/prevención & control , Huesos/fisiopatología , Músculo Esquelético/fisiopatología , Atrofia Muscular/prevención & control , Simulación de Ingravidez/efectos adversos , Animales , Fenómenos Biomecánicos , Resorción Ósea/etiología , Huesos/patología , Femenino , Gravitación , Marte , Ratones , Ratones Endogámicos BALB C , Luna , Atrofia Muscular/etiología , Osteogénesis , Pérdida de Peso , Soporte de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA