Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 183(5): 1162-1184, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33242416

RESUMEN

Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.


Asunto(s)
Medio Ambiente Extraterrestre , Vuelo Espacial , Astronautas , Salud , Humanos , Microbiota , Factores de Riesgo
3.
Nature ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862028

RESUMEN

Spaceflight induces molecular, cellular, and physiological shifts in astronauts and poses myriad biomedical challenges to the human body, which are becoming increasingly relevant as more humans venture into space1-6. Yet, current frameworks for aerospace medicine are nascent and lag far behind advancements in precision medicine on Earth, underscoring the need for rapid development of space medicine databases, tools, and protocols. Here, we present the Space Omics and Medical Atlas (SOMA), an integrated data and sample repository for clinical, cellular, and multi-omic research profiles from a diverse range of missions, including the NASA Twins Study7, JAXA CFE study8,9, SpaceX Inspiration4 crew10-12, plus Axiom and Polaris. The SOMA resource represents a >10-fold increase in publicly available human space omics data, with matched samples available from the Cornell Aerospace Medicine Biobank. The Atlas includes extensive molecular and physiological profiles encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and microbiome data sets, which reveal some consistent features across missions, including cytokine shifts, telomere elongation, and gene expression changes, as well as mission-specific molecular responses and links to orthologous, tissue-specific murine data sets. Leveraging the datasets, tools, and resources in SOMA can help accelerate precision aerospace medicine, bringing needed health monitoring, risk mitigation, and countermeasures data for upcoming lunar, Mars, and exploration-class missions.

4.
Proc Natl Acad Sci U S A ; 114(40): 10719-10724, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28893994

RESUMEN

There is emerging evidence that the commensal microbiota has a role in the pathogenesis of multiple sclerosis (MS), a putative autoimmune disease of the CNS. Here, we compared the gut microbial composition of 34 monozygotic twin pairs discordant for MS. While there were no major differences in the overall microbial profiles, we found a significant increase in some taxa such as Akkermansia in untreated MS twins. Furthermore, most notably, when transplanted to a transgenic mouse model of spontaneous brain autoimmunity, MS twin-derived microbiota induced a significantly higher incidence of autoimmunity than the healthy twin-derived microbiota. The microbial profiles of the colonized mice showed a high intraindividual and remarkable temporal stability with several differences, including Sutterella, an organism shown to induce a protective immunoregulatory profile in vitro. Immune cells from mouse recipients of MS-twin samples produced less IL-10 than immune cells from mice colonized with healthy-twin samples. IL-10 may have a regulatory role in spontaneous CNS autoimmunity, as neutralization of the cytokine in mice colonized with healthy-twin fecal samples increased disease incidence. These findings provide evidence that MS-derived microbiota contain factors that precipitate an MS-like autoimmune disease in a transgenic mouse model. They hence encourage the detailed search for protective and pathogenic microbial components in human MS.


Asunto(s)
Encéfalo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Animales , Encéfalo/microbiología , Encéfalo/patología , Estudios de Cohortes , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Masculino , Metagenómica , Ratones , Persona de Mediana Edad , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Adulto Joven
5.
Proc Natl Acad Sci U S A ; 114(40): 10713-10718, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28893978

RESUMEN

The gut microbiota regulates T cell functions throughout the body. We hypothesized that intestinal bacteria impact the pathogenesis of multiple sclerosis (MS), an autoimmune disorder of the CNS and thus analyzed the microbiomes of 71 MS patients not undergoing treatment and 71 healthy controls. Although no major shifts in microbial community structure were found, we identified specific bacterial taxa that were significantly associated with MS. Akkermansia muciniphila and Acinetobacter calcoaceticus, both increased in MS patients, induced proinflammatory responses in human peripheral blood mononuclear cells and in monocolonized mice. In contrast, Parabacteroides distasonis, which was reduced in MS patients, stimulated antiinflammatory IL-10-expressing human CD4+CD25+ T cells and IL-10+FoxP3+ Tregs in mice. Finally, microbiota transplants from MS patients into germ-free mice resulted in more severe symptoms of experimental autoimmune encephalomyelitis and reduced proportions of IL-10+ Tregs compared with mice "humanized" with microbiota from healthy controls. This study identifies specific human gut bacteria that regulate adaptive autoimmune responses, suggesting therapeutic targeting of the microbiota as a treatment for MS.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal , Leucocitos Mononucleares/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T/inmunología , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/microbiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Leucocitos Mononucleares/microbiología , Leucocitos Mononucleares/patología , Masculino , Ratones , Esclerosis Múltiple/microbiología , Esclerosis Múltiple/patología , Linfocitos T/microbiología , Linfocitos T/patología
6.
Int J Mol Sci ; 19(11)2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30463349

RESUMEN

In preparation for lunar and Mars missions it is essential to consider the challenges to human health that are posed by long-duration deep space habitation via multiple stressors, including ionizing radiation, gravitational changes during flight and in orbit, other aspects of the space environment such as high level of carbon dioxide, and psychological stress from confined environment and social isolation. It remains unclear how these stressors individually or in combination impact the central nervous system (CNS), presenting potential obstacles for astronauts engaged in deep space travel. Although human spaceflight research only within the last decade has started to include the effects of radiation transmitted by galactic cosmic rays to the CNS, radiation is currently considered to be one of the main stressors for prolonged spaceflight and deep space exploration. Here we will review the current knowledge of CNS damage caused by simulated space radiation with an emphasis on neuronal and glial responses along with cognitive functions. Furthermore, we will present novel experimental approaches to integrate the knowledge into more comprehensive studies, including multiple stressors at once and potential translation to human functions. Finally, we will discuss the need for developing biomarkers as predictors for cognitive decline and therapeutic countermeasures to prevent CNS damage and the loss of cognitive abilities.


Asunto(s)
Sistema Nervioso Central/fisiología , Sistema Nervioso Central/efectos de la radiación , Radiación Cósmica , Animales , Conducta Animal/efectos de la radiación , Sistema Nervioso Central/patología , Cognición , Humanos , Inflamación/patología , Neuronas/patología
7.
J Immunol ; 193(1): 139-49, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24860191

RESUMEN

The balance between controlling infection and limiting inflammation is particularly precarious in the brain because of its unique vulnerability to the toxic effects of inflammation. Astrocytes have been implicated as key regulators of neuroinflammation in CNS infections, including infection with Toxoplasma gondii, a protozoan parasite that naturally establishes a chronic CNS infection in mice and humans. In CNS toxoplasmosis, astrocytes are critical to controlling parasite growth. They secrete proinflammatory cytokines and physically encircle parasites. However, the molecular mechanisms used by astrocytes to limit neuroinflammation during toxoplasmic encephalitis have not yet been identified. TGF-ß signaling in astrocytes is of particular interest because TGF-ß is universally upregulated during CNS infection and serves master regulatory and primarily anti-inflammatory functions. We report in this study that TGF-ß signaling is activated in astrocytes during toxoplasmic encephalitis and that inhibition of astrocytic TGF-ß signaling increases immune cell infiltration, uncouples proinflammatory cytokine and chemokine production from CNS parasite burden, and increases neuronal injury. Remarkably, we show that the effects of inhibiting astrocytic TGF-ß signaling are independent of parasite burden and the ability of GFAP(+) astrocytes to physically encircle parasites.


Asunto(s)
Astrocitos/inmunología , Neuronas/inmunología , Transducción de Señal/inmunología , Toxoplasma/inmunología , Toxoplasmosis Cerebral/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Astrocitos/parasitología , Astrocitos/patología , Quimiocinas/genética , Quimiocinas/inmunología , Proteína Ácida Fibrilar de la Glía , Humanos , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neuronas/parasitología , Neuronas/patología , Transducción de Señal/genética , Toxoplasma/genética , Toxoplasmosis Cerebral/genética , Toxoplasmosis Cerebral/patología , Factor de Crecimiento Transformador beta/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
8.
J Neurosci ; 33(40): 16016-32, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-24089506

RESUMEN

Prostaglandin E2 (PGE2), a potent lipid signaling molecule, modulates inflammatory responses through activation of downstream G-protein coupled EP(1-4) receptors. Here, we investigated the cell-specific in vivo function of PGE2 signaling through its E-prostanoid 2 (EP2) receptor in murine innate immune responses systemically and in the CNS. In vivo, systemic administration of lipopolysaccharide (LPS) resulted in a broad induction of cytokines and chemokines in plasma that was significantly attenuated in EP2-deficient mice. Ex vivo stimulation of peritoneal macrophages with LPS elicited proinflammatory responses that were dependent on EP2 signaling and that overlapped with in vivo plasma findings, suggesting that myeloid-lineage EP2 signaling is a major effector of innate immune responses. Conditional deletion of the EP2 receptor in myeloid lineage cells in Cd11bCre;EP2(lox/lox) mice attenuated plasma inflammatory responses and transmission of systemic inflammation to the brain was inhibited, with decreased hippocampal inflammatory gene expression and cerebral cortical levels of IL-6. Conditional deletion of EP2 significantly blunted microglial and astrocytic inflammatory responses to the neurotoxin MPTP and reduced striatal dopamine turnover. Suppression of microglial EP2 signaling also increased numbers of dopaminergic (DA) neurons in the substantia nigra independent of MPTP treatment, suggesting that microglial EP2 may influence development or survival of DA neurons. Unbiased microarray analysis of microglia isolated from adult Cd11bCre;EP2(lox/lox) and control mice demonstrated a broad downregulation of inflammatory pathways with ablation of microglial EP2 receptor. Together, these data identify a cell-specific proinflammatory role for macrophage/microglial EP2 signaling in innate immune responses systemically and in brain.


Asunto(s)
Encéfalo/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Encéfalo/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Inflamación/inducido químicamente , Inflamación/genética , Lipopolisacáridos , Macrófagos/efectos de los fármacos , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Subtipo EP2 de Receptores de Prostaglandina E/genética
9.
Glia ; 62(8): 1227-40, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24733756

RESUMEN

Astrocytes limit inflammation after CNS injury, at least partially by physically containing it within an astrocytic scar at the injury border. We report here that astrocytic transforming growth factor-beta (TGFß) signaling is a second, distinct mechanism that astrocytes utilize to limit neuroinflammation. TGFßs are anti-inflammatory and neuroprotective cytokines that are upregulated subacutely after stroke, during a clinically accessible time window. We have previously demonstrated that TGFßs signal to astrocytes, neurons and microglia in the stroke border days after stroke. To investigate whether TGFß affects astrocyte immunoregulatory functions, we engineered "Ast-Tbr2DN" mice where TGFß signaling is inhibited specifically in astrocytes. Despite having a similar infarct size to wildtype controls, Ast-Tbr2DN mice exhibited significantly more neuroinflammation during the subacute period after distal middle cerebral occlusion (dMCAO) stroke. The peri-infarct cortex of Ast-Tbr2DN mice contained over 60% more activated CD11b(+) monocytic cells and twice as much immunostaining for the activated microglia and macrophage marker CD68 than controls. Astrocytic scarring was not altered in Ast-Tbr2DN mice. However, Ast-Tbr2DN mice were unable to upregulate TGF-ß1 and its activator thrombospondin-1 2 days after dMCAO. As a result, the normal upregulation of peri-infarct TGFß signaling was blunted in Ast-Tbr2DN mice. In this setting of lower TGFß signaling and excessive neuroinflammation, we observed worse motor outcomes and late infarct expansion after photothrombotic motor cortex stroke. Taken together, these data demonstrate that TGFß signaling is a molecular mechanism by which astrocytes limit neuroinflammation, activate TGFß in the peri-infarct cortex and preserve brain function during the subacute period after stroke.


Asunto(s)
Astrocitos/inmunología , Corteza Cerebral/inmunología , Infarto de la Arteria Cerebral Media/inmunología , Accidente Cerebrovascular/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Astrocitos/patología , Antígeno CD11b/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Infarto de la Arteria Cerebral Media/patología , Macrófagos/patología , Macrófagos/fisiología , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Monocitos/patología , Monocitos/fisiología , Actividad Motora/fisiología , Neuroinmunomodulación/fisiología , Transducción de Señal , Accidente Cerebrovascular/patología , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/metabolismo
10.
NPJ Microgravity ; 10(1): 63, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862517

RESUMEN

Spaceflight and terrestrial spaceflight analogs can alter immune phenotypes. Macrophages are important immune cells that bridge the innate and adaptive immune systems and participate in immunoregulatory processes of homeostasis. Furthermore, macrophages are critically involved in initiating immunity, defending against injury and infection, and are also involved in immune resolution and wound healing. Heterogeneous populations of macrophage-type cells reside in many tissues and cause a variety of tissue-specific effects through direct or indirect interactions with other physiological systems, including the nervous and endocrine systems. It is vital to understand how macrophages respond to the unique environment of space to safeguard crew members with appropriate countermeasures for future missions in low Earth orbit and beyond. This review highlights current literature on macrophage responses to spaceflight and spaceflight analogs.

11.
Nat Commun ; 15(1): 4778, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862479

RESUMEN

Impairment of the central nervous system (CNS) poses a significant health risk for astronauts during long-duration space missions. In this study, we employed an innovative approach by integrating single-cell multiomics (transcriptomics and chromatin accessibility) with spatial transcriptomics to elucidate the impact of spaceflight on the mouse brain in female mice. Our comparative analysis between ground control and spaceflight-exposed animals revealed significant alterations in essential brain processes including neurogenesis, synaptogenesis and synaptic transmission, particularly affecting the cortex, hippocampus, striatum and neuroendocrine structures. Additionally, we observed astrocyte activation and signs of immune dysfunction. At the pathway level, some spaceflight-induced changes in the brain exhibit similarities with neurodegenerative disorders, marked by oxidative stress and protein misfolding. Our integrated spatial multiomics approach serves as a stepping stone towards understanding spaceflight-induced CNS impairments at the level of individual brain regions and cell types, and provides a basis for comparison in future spaceflight studies. For broader scientific impact, all datasets from this study are available through an interactive data portal, as well as the National Aeronautics and Space Administration (NASA) Open Science Data Repository (OSDR).


Asunto(s)
Encéfalo , Neuronas , Vuelo Espacial , Animales , Ratones , Femenino , Encéfalo/metabolismo , Encéfalo/patología , Neuronas/metabolismo , Transcriptoma , Neurogénesis , Análisis de la Célula Individual , Ratones Endogámicos C57BL , Transmisión Sináptica , Ingravidez/efectos adversos , Astrocitos/metabolismo , Estrés Oxidativo , Perfilación de la Expresión Génica , Multiómica
12.
Life Sci Space Res (Amst) ; 36: 47-58, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36682829

RESUMEN

Exposure to ionizing radiation is considered by NASA to be a major health hazard for deep space exploration missions. Ionizing radiation sensitivity is modulated by both genomic and environmental factors. Understanding their contributions is crucial for designing experiments in model organisms, evaluating the risk of deep space (i.e. high-linear energy transfer, or LET, particle) radiation exposure in astronauts, and also selecting therapeutic irradiation regimes for cancer patients. We identified single nucleotide polymorphisms in 15 strains of mice, including 10 collaborative cross model strains and 5 founder strains, associated with spontaneous and ionizing radiation-induced in vitro DNA damage quantified based on immunofluorescent tumor protein p53 binding protein (53BP1) positive nuclear foci. Statistical analysis suggested an association with pathways primarily related to cellular signaling, metabolism, tumorigenesis and nervous system damage. We observed different genomic associations in early (4 and 8 h) responses to different LET radiation, while later (24 hour) DNA damage responses showed a stronger overlap across all LETs. Furthermore, a subset of pathways was associated with spontaneous DNA damage, suggesting 53BP1 positive foci as a potential biomarker for DNA integrity in mouse models. Our results suggest several mouse strains as new models to further study the impact of ionizing radiation and validate the identified genetic loci. We also highlight the importance of future human in vitro studies to refine the association of genes and pathways with the DNA damage response to ionizing radiation and identify targets for space travel countermeasures.


Asunto(s)
Daño del ADN , Neoplasias , Humanos , Ratones , Animales , Reparación del ADN , Radiación Ionizante , Genómica
13.
Stroke ; 43(7): 1918-24, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535263

RESUMEN

BACKGROUND AND PURPOSE: Stroke is the leading cause of long-term disability in the United States, yet no drugs are available that are proven to improve recovery. Brain-derived neurotrophic factor stimulates neurogenesis and plasticity, processes that are implicated in stroke recovery. It binds to both the tropomyosin-related kinase B and p75 neurotrophin receptors. However, brain-derived neurotrophic factor is not a feasible therapeutic agent, and no small molecule exists that can reproduce its binding to both receptors. We tested the hypothesis that a small molecule (LM22A-4) that selectively targets tropomyosin-related kinase B would promote neurogenesis and functional recovery after stroke. METHODS: Four-month-old mice were trained on motor tasks before stroke. After stroke, functional test results were used to randomize mice into 2 equally, and severely, impaired groups. Beginning 3 days after stroke, mice received LM22A-4 or saline vehicle daily for 10 weeks. RESULTS: LM22A-4 treatment significantly improved limb swing speed and accelerated the return to normal gait accuracy after stroke. LM22A-4 treatment also doubled both the number of new mature neurons and immature neurons adjacent to the stroke. Drug-induced differences were not observed in angiogenesis, dendritic arborization, axonal sprouting, glial scar formation, or neuroinflammation. CONCLUSIONS: A small molecule agonist of tropomyosin-related kinase B improves functional recovery from stroke and increases neurogenesis when administered beginning 3 days after stroke. These findings provide proof-of-concept that targeting of tropomyosin-related kinase B alone is capable of promoting one or more mechanisms relevant to stroke recovery. LM22A-4 or its derivatives might therefore serve as "pro-recovery" therapeutic agents for stroke.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Recuperación de la Función/efectos de los fármacos , Tropomiosina/administración & dosificación , Animales , Hipoxia-Isquemia Encefálica/fisiopatología , Ligandos , Masculino , Glicoproteínas de Membrana/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Proteínas Tirosina Quinasas/uso terapéutico , Distribución Aleatoria , Recuperación de la Función/fisiología , Accidente Cerebrovascular , Tropomiosina/química
14.
Front Immunol ; 13: 864923, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275678

RESUMEN

Central nervous system (CNS) damage by galactic cosmic ray radiation is a major health risk for human deep space exploration. Simulated galactic cosmic rays or their components, especially high Z-high energy particles such as 56Fe ions, cause neurodegeneration and neuroinflammation in rodent models. CNS damage can be partially mediated by the blood-brain barrier, which regulates systemic interactions between CNS and the rest of the body. Astrocytes are major cellular regulators of blood-brain barrier permeability that also modulate neuroinflammation and neuronal health. However, astrocyte roles in regulating CNS and blood-brain barrier responses to space radiation remain little understood, especially in human tissue analogs. In this work, we used a novel high-throughput human organ-on-a-chip system to evaluate blood-brain barrier impairments and astrocyte functions 1-7 days after exposure to 600 MeV/n 56Fe particles and simplified simulated galactic cosmic rays. We show that simulated deep space radiation causes vascular permeability, oxidative stress, inflammation and delayed astrocyte activation in a pattern resembling CNS responses to brain injury. Furthermore, our results indicate that astrocytes have a dual role in regulating radiation responses: they exacerbate blood-brain barrier permeability acutely after irradiation, followed by switching to a more protective phenotype by reducing oxidative stress and pro-inflammatory cytokine and chemokine secretion during the subacute stage.


Asunto(s)
Astrocitos , Dispositivos Laboratorio en un Chip , Humanos , Iones , Citocinas , Quimiocinas
15.
NAR Cancer ; 3(4): zcab046, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35692378

RESUMEN

Radiation-induced foci (RIF) are nuclear puncta visualized by immunostaining of proteins that regulate DNA double-strand break (DSB) repair after exposure to ionizing radiation. RIF are a standard metric for measuring DSB formation and repair in clinical, environmental and space radiobiology. The time course and dose dependence of their formation has great potential to predict in vivo responses to ionizing radiation, predisposition to cancer and probability of adverse reactions to radiotherapy. However, increasing complexity of experimentally and therapeutically setups (charged particle, FLASH …) is associated with several confounding factors that must be taken into account when interpreting RIF values. In this review, we discuss the spatiotemporal characteristics of RIF development after irradiation, addressing the common confounding factors, including cell proliferation and foci merging. We also describe the relevant endpoints and mathematical models that enable accurate biological interpretation of RIF formation and resolution. Finally, we discuss the use of RIF as a biomarker for quantification and prediction of in vivo radiation responses, including important caveats relating to the choice of the biological endpoint and the detection method. This review intends to help scientific community design radiobiology experiments using RIF as a key metric and to provide suggestions for their biological interpretation.

16.
Int J Radiat Biol ; 97(sup1): S132-S150, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32946305

RESUMEN

PURPOSE: Harmful effects of ionizing radiation on the Central Nervous System (CNS) are a concerning outcome in the field of cancer radiotherapy and form a major risk for deep space exploration. Both acute and chronic CNS irradiation induce a complex network of molecular and cellular alterations including DNA damage, oxidative stress, cell death and systemic inflammation, leading to changes in neuronal structure and synaptic plasticity with behavioral and cognitive consequences in animal models. Due to this complexity, countermeasure or therapeutic approaches to reduce the harmful effects of ionizing radiation include a wide range of protective and mitigative strategies, which merit a thorough comparative analysis. MATERIALS AND METHODS: We reviewed current approaches for developing countermeasures to both targeted and non-targeted effects of ionizing radiation on the CNS from the molecular and cellular to the behavioral level. RESULTS: We focus on countermeasures that aim to mitigate the four main detrimental actions of radiation on CNS: DNA damage, free radical formation and oxidative stress, cell death, and harmful systemic responses including tissue death and neuroinflammation. We propose a comprehensive review of CNS radiation countermeasures reported for the full range of irradiation types (photons and particles, low and high linear energy transfer) and doses (from a fraction of gray to several tens of gray, fractionated and unfractionated), with a particular interest for exposure conditions relevant to deep-space environment and radiotherapy. Our review reveals the importance of combined strategies that increase DNA protection and repair, reduce free radical formation and increase their elimination, limit inflammation and improve cell viability, limit tissue damage and increase repair and plasticity. CONCLUSIONS: The majority of therapeutic approaches to protect the CNS from ionizing radiation have been limited to acute high dose and high dose rate gamma irradiation, and few are translatable from animal models to potential human application due to harmful side effects and lack of blood-brain barrier permeability that precludes peripheral administration. Therefore, a promising research direction would be to focus on practical applicability and effectiveness in a wider range of irradiation paradigms, from fractionated therapeutic to deep space radiation. In addition to discovering novel therapeutics, it would be worth maximizing the benefits and reducing side effects of those that already exist. Finally, we suggest that novel cellular and tissue models for developing and testing countermeasures in the context of other impairments might also be applied to the field of CNS responses to ionizing radiation.


Asunto(s)
Radiación Ionizante , Roedores , Animales , Sistema Nervioso Central , Radicales Libres , Inflamación
17.
Life (Basel) ; 11(1)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445483

RESUMEN

There has long been an interest in understanding how the hazards from spaceflight may trigger or exacerbate human diseases. With the goal of advancing our knowledge on physiological changes during space travel, NASA GeneLab provides an open-source repository of multi-omics data from real and simulated spaceflight studies. Alone, this data enables identification of biological changes during spaceflight, but cannot infer how that may impact an astronaut at the phenotypic level. To bridge this gap, Scalable Precision Medicine Oriented Knowledge Engine (SPOKE), a heterogeneous knowledge graph connecting biological and clinical data from over 30 databases, was used in combination with GeneLab transcriptomic data from six studies. This integration identified critical symptoms and physiological changes incurred during spaceflight.

18.
iScience ; 24(4): 102361, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33870146

RESUMEN

With the development of transcriptomic technologies, we are able to quantify precise changes in gene expression profiles from astronauts and other organisms exposed to spaceflight. Members of NASA GeneLab and GeneLab-associated analysis working groups (AWGs) have developed a consensus pipeline for analyzing short-read RNA-sequencing data from spaceflight-associated experiments. The pipeline includes quality control, read trimming, mapping, and gene quantification steps, culminating in the detection of differentially expressed genes. This data analysis pipeline and the results of its execution using data submitted to GeneLab are now all publicly available through the GeneLab database. We present here the full details and rationale for the construction of this pipeline in order to promote transparency, reproducibility, and reusability of pipeline data; to provide a template for data processing of future spaceflight-relevant datasets; and to encourage cross-analysis of data from other databases with the data available in GeneLab.

19.
J Neuroinflammation ; 7: 62, 2010 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-20937129

RESUMEN

BACKGROUND: TGFß is both neuroprotective and a key immune system modulator and is likely to be an important target for future stroke therapy. The precise function of increased TGF-ß1 after stroke is unknown and its pleiotropic nature means that it may convey a neuroprotective signal, orchestrate glial scarring or function as an important immune system regulator. We therefore investigated the time course and cell-specificity of TGFß signaling after stroke, and whether its signaling pattern is altered by gender and aging. METHODS: We performed distal middle cerebral artery occlusion strokes on 5 and 18 month old TGFß reporter mice to get a readout of TGFß responses after stroke in real time. To determine which cell type is the source of increased TGFß production after stroke, brain sections were stained with an anti-TGFß antibody, colocalized with markers for reactive astrocytes, neurons, and activated microglia. To determine which cells are responding to TGFß after stroke, brain sections were double-labelled with anti-pSmad2, a marker of TGFß signaling, and markers of neurons, oligodendrocytes, endothelial cells, astrocytes and microglia. RESULTS: TGFß signaling increased 2 fold after stroke, beginning on day 1 and peaking on day 7. This pattern of increase was preserved in old animals and absolute TGFß signaling in the brain increased with age. Activated microglia and macrophages were the predominant source of increased TGFß after stroke and astrocytes and activated microglia and macrophages demonstrated dramatic upregulation of TGFß signaling after stroke. TGFß signaling in neurons and oligodendrocytes did not undergo marked changes. CONCLUSIONS: We found that TGFß signaling increases with age and that astrocytes and activated microglia and macrophages are the main cell types that undergo increased TGFß signaling in response to post-stroke increases in TGFß. Therefore increased TGFß after stroke likely regulates glial scar formation and the immune response to stroke.


Asunto(s)
Envejecimiento/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Macrófagos/metabolismo , Accidente Cerebrovascular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Envejecimiento/inmunología , Análisis de Varianza , Animales , Astrocitos/inmunología , Western Blotting , Encéfalo/inmunología , Femenino , Técnica del Anticuerpo Fluorescente , Macrófagos/inmunología , Masculino , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Transducción de Señal , Accidente Cerebrovascular/inmunología , Factor de Crecimiento Transformador beta/inmunología
20.
Radiat Res ; 194(5): 485-499, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32991727

RESUMEN

We present a novel mathematical formalism to predict the kinetics of DNA damage repair after exposure to both low- and high-LET radiation (X rays; 350 MeV/n 40Ar; 600 MeV/n 56Fe). Our method is based on monitoring DNA damage repair protein 53BP1 that forms radiation-induced foci (RIF) at locations of DNA double-strand breaks (DSB) in the nucleus and comparing its expression in primary skin fibroblasts isolated from 15 mice strains. We previously reported strong evidence for clustering of nearby DSB into single repair units as opposed to the classic "contact-first" model where DSB are considered immobile. Here we apply this clustering model to evaluate the number of remaining RIF over time. We also show that the newly introduced kinetic metrics can be used as surrogate biomarkers for in vivo radiation toxicity, with potential applications in radiotherapy and human space exploration. In particular, we observed an association between the characteristic time constant of RIF repair measured in vitro and survival levels of immune cells collected from irradiated mice. Moreover, the speed of DNA damage repair correlated not only with radiation-induced cellular survival in vivo, but also with spontaneous cancer incidence data collected from the Mouse Tumor Biology database, suggesting a relationship between the efficiency of DSB repair after irradiation and cancer risk.


Asunto(s)
Reparación del ADN , ADN/efectos de la radiación , Ratones Endogámicos/genética , Tolerancia a Radiación/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Medicina Aeroespacial , Animales , Células Cultivadas , ADN/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN , Femenino , Fibroblastos/efectos de la radiación , Iones Pesados , Incidencia , Cinética , Transferencia Lineal de Energía , Masculino , Ratones , Modelos Genéticos , Neoplasias/epidemiología , Neoplasias/genética , Neoplasias/veterinaria , Exposición a la Radiación , Efectividad Biológica Relativa , Riesgo , Enfermedades de los Roedores/epidemiología , Enfermedades de los Roedores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA