Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 183(5): 1162-1184, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242416

ABSTRACT

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.


Subject(s)
Extraterrestrial Environment , Space Flight , Astronauts , Health , Humans , Microbiota , Risk Factors
2.
Cell ; 183(5): 1185-1201.e20, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242417

ABSTRACT

Spaceflight is known to impose changes on human physiology with unknown molecular etiologies. To reveal these causes, we used a multi-omics, systems biology analytical approach using biomedical profiles from fifty-nine astronauts and data from NASA's GeneLab derived from hundreds of samples flown in space to determine transcriptomic, proteomic, metabolomic, and epigenetic responses to spaceflight. Overall pathway analyses on the multi-omics datasets showed significant enrichment for mitochondrial processes, as well as innate immunity, chronic inflammation, cell cycle, circadian rhythm, and olfactory functions. Importantly, NASA's Twin Study provided a platform to confirm several of our principal findings. Evidence of altered mitochondrial function and DNA damage was also found in the urine and blood metabolic data compiled from the astronaut cohort and NASA Twin Study data, indicating mitochondrial stress as a consistent phenotype of spaceflight.


Subject(s)
Genomics , Mitochondria/pathology , Space Flight , Stress, Physiological , Animals , Circadian Rhythm , Extracellular Matrix/metabolism , Humans , Immunity, Innate , Lipid Metabolism , Metabolic Flux Analysis , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscles/immunology , Organ Specificity , Smell/physiology
4.
Nature ; 632(8027): 995-1008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862027

ABSTRACT

The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.


Subject(s)
Aerospace Medicine , Astronauts , Multiomics , Space Flight , Humans , Aerospace Medicine/methods , Aerospace Medicine/trends , Biological Specimen Banks , Biomarkers/metabolism , Biomarkers/analysis , Cognition , Internationality , Monitoring, Physiologic/methods , Monitoring, Physiologic/trends , Multiomics/methods , Multiomics/trends , Pharmacogenetics/methods , Pharmacogenetics/trends , Precision Medicine/methods , Precision Medicine/trends , Space Flight/methods , Space Flight/trends
5.
Cell ; 144(5): 732-44, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21353298

ABSTRACT

Double-strand breaks (DSBs) in heterochromatic repetitive DNAs pose significant threats to genome integrity, but information about how such lesions are processed and repaired is sparse. We observe dramatic expansion and dynamic protrusions of the heterochromatin domain in response to ionizing radiation (IR) in Drosophila cells. We also find that heterochromatic DSBs are repaired by homologous recombination (HR) but with striking differences from euchromatin. Proteins involved in early HR events (resection) are rapidly recruited to DSBs within heterochromatin. In contrast, Rad51, which mediates strand invasion, only associates with DSBs that relocalize outside of the domain. Heterochromatin expansion and relocalization of foci require checkpoint and resection proteins. Finally, the Smc5/6 complex is enriched in heterochromatin and is required to exclude Rad51 from the domain and prevent abnormal recombination. We propose that the spatial and temporal control of DSB repair in heterochromatin safeguards genome stability by preventing aberrant exchanges between repeats.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Repair , Drosophila Proteins/metabolism , Recombination, Genetic , Animals , Cell Cycle Proteins/metabolism , Chromobox Protein Homolog 5 , DNA Breaks, Double-Stranded , Drosophila melanogaster/metabolism , Heterochromatin , Rad51 Recombinase/metabolism
6.
Bioinformatics ; 39(2)2023 02 03.
Article in English | MEDLINE | ID: mdl-36759942

ABSTRACT

MOTIVATION: Knowledge graphs (KGs) are being adopted in industry, commerce and academia. Biomedical KG presents a challenge due to the complexity, size and heterogeneity of the underlying information. RESULTS: In this work, we present the Scalable Precision Medicine Open Knowledge Engine (SPOKE), a biomedical KG connecting millions of concepts via semantically meaningful relationships. SPOKE contains 27 million nodes of 21 different types and 53 million edges of 55 types downloaded from 41 databases. The graph is built on the framework of 11 ontologies that maintain its structure, enable mappings and facilitate navigation. SPOKE is built weekly by python scripts which download each resource, check for integrity and completeness, and then create a 'parent table' of nodes and edges. Graph queries are translated by a REST API and users can submit searches directly via an API or a graphical user interface. Conclusions/Significance: SPOKE enables the integration of seemingly disparate information to support precision medicine efforts. AVAILABILITY AND IMPLEMENTATION: The SPOKE neighborhood explorer is available at https://spoke.rbvi.ucsf.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Pattern Recognition, Automated , Precision Medicine , Databases, Factual
7.
Nucleic Acids Res ; 49(D1): D1515-D1522, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33080015

ABSTRACT

The mission of NASA's GeneLab database (https://genelab.nasa.gov/) is to collect, curate, and provide access to the genomic, transcriptomic, proteomic and metabolomic (so-called 'omics') data from biospecimens flown in space or exposed to simulated space stressors, maximizing their utilization. This large collection of data enables the exploration of molecular network responses to space environments using a systems biology approach. We review here the various components of the GeneLab platform, including the new data repository web interface, and the GeneLab Online Data Entry (GEODE) web portal, which will support the expansion of the database in the future to include companion non-omics assay data. We discuss our design for GEODE, particularly how it promotes investigators providing more accurate metadata, reducing the curation effort required of GeneLab staff. We also introduce here a new GeneLab Application Programming Interface (API) specifically designed to support tools for the visualization of processed omics data. We review the outreach efforts by GeneLab to utilize the spaceflight data in the repository to generate novel discoveries and develop new hypotheses, including spearheading data analysis working groups, and a high school student training program. All these efforts are aimed ultimately at supporting precision risk management for human space exploration.


Subject(s)
Databases, Genetic , Genome , Software , Weightlessness , Animals , Astronauts , Bacteria/genetics , Bacteria/metabolism , Fishes/genetics , Fishes/metabolism , Gene Expression Regulation , Humans , Information Dissemination , Insecta/genetics , Insecta/metabolism , Internet , Mice , Nematoda/genetics , Nematoda/metabolism , Plants/genetics , Plants/metabolism , Space Flight , Weightlessness Simulation
8.
Nucleic Acids Res ; 49(W1): W114-W124, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34037798

ABSTRACT

In molecular biology and genetics, there is a large gap between the ease of data collection and our ability to extract knowledge from these data. Contributing to this gap is the fact that living organisms are complex systems whose emerging phenotypes are the results of multiple complex interactions taking place on various pathways. This demands powerful yet user-friendly pathway analysis tools to translate the now abundant high-throughput data into a better understanding of the underlying biological phenomena. Here we introduce Consensus Pathway Analysis (CPA), a web-based platform that allows researchers to (i) perform pathway analysis using eight established methods (GSEA, GSA, FGSEA, PADOG, Impact Analysis, ORA/Webgestalt, KS-test, Wilcox-test), (ii) perform meta-analysis of multiple datasets, (iii) combine methods and datasets to accurately identify the impacted pathways underlying the studied condition and (iv) interactively explore impacted pathways, and browse relationships between pathways and genes. The platform supports three types of input: (i) a list of differentially expressed genes, (ii) genes and fold changes and (iii) an expression matrix. It also allows users to import data from NCBI GEO. The CPA platform currently supports the analysis of multiple organisms using KEGG and Gene Ontology, and it is freely available at http://cpa.tinnguyen-lab.com.


Subject(s)
Gene Expression , Software , Alzheimer Disease/genetics , Datasets as Topic , Gene Ontology , Humans , Internet
9.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502375

ABSTRACT

Bioinformatics approaches have proven useful in understanding biological responses to spaceflight. Spaceflight experiments remain resource intensive and rare. One outstanding issue is how to maximize scientific output from a limited number of omics datasets from traditional animal models including nematodes, fruitfly, and rodents. The utility of omics data from invertebrate models in anticipating mammalian responses to spaceflight has not been fully explored. Hence, we performed comparative analyses of transcriptomes of soleus and extensor digitorum longus (EDL) in mice that underwent 37 days of spaceflight. Results indicate shared stress responses and altered circadian rhythm. EDL showed more robust growth signals and Pde2a downregulation, possibly underlying its resistance to atrophy versus soleus. Spaceflight and hindlimb unloading mice shared differential regulation of proliferation, circadian, and neuronal signaling. Shared gene regulation in muscles of humans on bedrest and space flown rodents suggest targets for mitigating muscle atrophy in space and on Earth. Spaceflight responses of C. elegans were more similar to EDL. Discrete life stages of D. melanogaster have distinct utility in anticipating EDL and soleus responses. In summary, spaceflight leads to shared and discrete molecular responses between muscle types and invertebrate models may augment mechanistic knowledge gained from rodent spaceflight and ground-based studies.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Weightlessness/adverse effects , Animals , Caenorhabditis elegans , Circadian Rhythm/physiology , Databases, Genetic , Drosophila melanogaster , Extraterrestrial Environment , Gene Expression/genetics , Gene Expression Profiling/methods , Hindlimb Suspension , Mice , Models, Animal , Space Flight , Stress, Physiological/physiology , Transcriptome/genetics
10.
Bioinformatics ; 35(10): 1753-1759, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30329036

ABSTRACT

MOTIVATION: To curate and organize expensive spaceflight experiments conducted aboard space stations and maximize the scientific return of investment, while democratizing access to vast amounts of spaceflight related omics data generated from several model organisms. RESULTS: The GeneLab Data System (GLDS) is an open access database containing fully coordinated and curated 'omics' (genomics, transcriptomics, proteomics, metabolomics) data, detailed metadata and radiation dosimetry for a variety of model organisms. GLDS is supported by an integrated data system allowing federated search across several public bioinformatics repositories. Archived datasets can be queried using full-text search (e.g. keywords, Boolean and wildcards) and results can be sorted in multifactorial manner using assistive filters. GLDS also provides a collaborative platform built on GenomeSpace for sharing files and analyses with collaborators. It currently houses 172 datasets and supports standard guidelines for submission of datasets, MIAME (for microarray), ENCODE Consortium Guidelines (for RNA-seq) and MIAPE Guidelines (for proteomics). AVAILABILITY AND IMPLEMENTATION: https://genelab.nasa.gov/.


Subject(s)
Space Flight , Computational Biology , Databases, Factual , Genomics
11.
Int J Mol Sci ; 20(3)2019 Feb 03.
Article in English | MEDLINE | ID: mdl-30717456

ABSTRACT

Space radiation has recently been considered a risk factor for astronauts' cardiac health. As an example, for the case of how to query and identify datasets within NASA's GeneLab database and demonstrate the database utility, we used an unbiased systems biology method for identifying key genes/drivers for the contribution of space radiation on the cardiovascular system. This knowledge can contribute to designing appropriate experiments targeting these specific pathways. Microarray data from cardiomyocytes of male C57BL/6 mice followed-up for 28 days after exposure to 900 mGy of 1 GeV proton or 150 mGy of 1 GeV/n 56Fe were compared to human endothelial cells (HUVECs) cultured for 7 days on the International Space Station (ISS). We observed common molecular pathways between simulated space radiation and HUVECs flown on the ISS. The analysis suggests FYN is the central driver/hub for the cardiovascular response to space radiation: the known oxidative stress induced immediately following radiation would only be transient and would upregulate FYN, which in turn would reduce reactive oxygen species (ROS) levels, protecting the cardiovascular system. The transcriptomic signature of exposure to protons was also much closer to the spaceflight signature than 56Fe's signature. To our knowledge, this is the first time GeneLab datasets were utilized to provide potential biological indications that the majority of ions on the ISS are protons, clearly illustrating the power of omics analysis. More generally, this work also demonstrates how to combine animal radiation studies done on the ground and spaceflight studies to evaluate human risk in space.


Subject(s)
Cardiovascular System/radiation effects , Myocytes, Cardiac/radiation effects , Proto-Oncogene Proteins c-fyn/genetics , Radiation, Ionizing , Space Flight , Transcriptome , Animals , Cardiovascular System/metabolism , Cells, Cultured , Cosmic Radiation , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-fyn/metabolism , Protons , Reactive Oxygen Species/metabolism
12.
Int J Mol Sci ; 19(11)2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30463349

ABSTRACT

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.


Subject(s)
Central Nervous System/physiology , Central Nervous System/radiation effects , Cosmic Radiation , Animals , Behavior, Animal/radiation effects , Central Nervous System/pathology , Cognition , Humans , Inflammation/pathology , Neurons/pathology
13.
Mol Carcinog ; 55(9): 1387-96, 2016 09.
Article in English | MEDLINE | ID: mdl-26310697

ABSTRACT

The tumor suppressor TP53 can initiate a plethora of anti-proliferative effects to maintain genomic integrity under conditions of genotoxic stress. The N-terminal proline-rich domain (PRD) of TP53 is important in the regulation of TP53 activity and stability. A common polymorphism at codon 72 in this region has been associated with altered cancer risk in humans. The Trp53ΔP mouse, which carries a germline homozygous deletion of a region of the PRD, does not develop spontaneous tumors in a mixed 129/Sv and C57BL/6 genetic background, but is highly susceptible to a broad range of tumor types following total body exposure to 4 Gy gamma (γ) radiation. This contrasts with the tumor spectrum in Trp53 null (-/-) mice, which mainly develop thymic lymphomas and osteosarcomas. Analysis of genomic instability in tissues and cells from Trp53ΔP mice demonstrated elevated basal levels of aneuploidy, but this is not sufficient to drive spontaneous tumorigenesis, which requires an additional DNA damage stimulus. Levels of genomic instability did not increase significantly in Trp53ΔP mice following irradiation exposure, suggesting that other radiation effects including tissue inflammation, altered metabolism or autophagy, may play an important role. The Trp53ΔP mouse is a novel model to dissect the mechanisms of tumor development induced by radiation exposure. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinogenesis/genetics , Genomic Instability , Neoplasms, Radiation-Induced/genetics , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Autophagy , Female , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Polyploidy , Proline/chemistry , Proline/genetics , Sequence Deletion , Tumor Suppressor Protein p53/chemistry
14.
Stem Cells ; 32(3): 649-61, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24038768

ABSTRACT

Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor ß (TGFß) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFß. Radiation-induced Notch concomitant with TGFß was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.


Subject(s)
Aging/pathology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/radiation effects , Mammary Neoplasms, Animal/pathology , Radiation, Ionizing , Receptors, Estrogen/metabolism , Stem Cells/pathology , Animals , Biomarkers/metabolism , Cell Line , Cell Lineage , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Computer Simulation , Dose-Response Relationship, Radiation , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Female , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Morphogenesis/drug effects , Morphogenesis/radiation effects , Receptors, Notch/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/radiation effects , Transforming Growth Factor beta/pharmacology
15.
PLoS Biol ; 10(12): e1001460, 2012.
Article in English | MEDLINE | ID: mdl-23300382

ABSTRACT

CENP-A (CID in flies) is the histone H3 variant essential for centromere specification, kinetochore formation, and chromosome segregation during cell division. Recent studies have elucidated major cell cycle mechanisms and factors critical for CENP-A incorporation in mitosis, predominantly in cultured cells. However, we do not understand the roles, regulation, and cell cycle timing of CENP-A assembly in somatic tissues in multicellular organisms and in meiosis, the specialized cell division cycle that gives rise to haploid gametes. Here we investigate the timing and requirements for CID assembly in mitotic tissues and male and female meiosis in Drosophila melanogaster, using fixed and live imaging combined with genetic approaches. We find that CID assembly initiates at late telophase and continues during G1 phase in somatic tissues in the organism, later than the metaphase assembly observed in cultured cells. Furthermore, CID assembly occurs at two distinct cell cycle phases during male meiosis: prophase of meiosis I and after exit from meiosis II, in spermatids. CID assembly in prophase I is also conserved in female meiosis. Interestingly, we observe a novel decrease in CID levels after the end of meiosis I and before meiosis II, which correlates temporally with changes in kinetochore organization and orientation. We also demonstrate that CID is retained on mature sperm despite the gross chromatin remodeling that occurs during protamine exchange. Finally, we show that the centromere proteins CAL1 and CENP-C are both required for CID assembly in meiosis and normal progression through spermatogenesis. We conclude that the cell cycle timing of CID assembly in meiosis is different from mitosis and that the efficient propagation of CID through meiotic divisions and on sperm is likely to be important for centromere specification in the developing zygote.


Subject(s)
Centromere/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Meiosis , Mitosis , Animals , Brain/cytology , Centromere Protein A , Chromosome Segregation , DNA-Binding Proteins/metabolism , Drosophila melanogaster/metabolism , Female , G1 Phase , Histones/metabolism , Larva/cytology , Male , Models, Biological , Protein Transport , Spermatids/cytology , Spermatids/metabolism , Telophase , Time Factors
16.
Proc Natl Acad Sci U S A ; 109(2): 443-8, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22184222

ABSTRACT

The concept of DNA "repair centers" and the meaning of radiation-induced foci (RIF) in human cells have remained controversial. RIFs are characterized by the local recruitment of DNA damage sensing proteins such as p53 binding protein (53BP1). Here, we provide strong evidence for the existence of repair centers. We used live imaging and mathematical fitting of RIF kinetics to show that RIF induction rate increases with increasing radiation dose, whereas the rate at which RIFs disappear decreases. We show that multiple DNA double-strand breaks (DSBs) 1 to 2 µm apart can rapidly cluster into repair centers. Correcting mathematically for the dose dependence of induction/resolution rates, we observe an absolute RIF yield that is surprisingly much smaller at higher doses: 15 RIF/Gy after 2 Gy exposure compared to approximately 64 RIF/Gy after 0.1 Gy. Cumulative RIF counts from time lapse of 53BP1-GFP in human breast cells confirmed these results. The standard model currently in use applies a linear scale, extrapolating cancer risk from high doses to low doses of ionizing radiation. However, our discovery of DSB clustering over such large distances casts considerable doubts on the general assumption that risk to ionizing radiation is proportional to dose, and instead provides a mechanism that could more accurately address risk dose dependency of ionizing radiation.


Subject(s)
Breast Neoplasms/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Intracellular Signaling Peptides and Proteins/metabolism , Cells, Cultured , DNA Repair/radiation effects , Female , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Models, Biological , Risk Assessment , Tumor Suppressor p53-Binding Protein 1
17.
PLoS Biol ; 9(11): e1001199, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22110403

ABSTRACT

Differentiated mammary epithelium shows apicobasal polarity, and loss of tissue organization is an early hallmark of breast carcinogenesis. In BRCA1 mutation carriers, accumulation of stem and progenitor cells in normal breast tissue and increased risk of developing tumors of basal-like type suggest that BRCA1 regulates stem/progenitor cell proliferation and differentiation. However, the function of BRCA1 in this process and its link to carcinogenesis remain unknown. Here we depict a molecular mechanism involving BRCA1 and RHAMM that regulates apicobasal polarity and, when perturbed, may increase risk of breast cancer. Starting from complementary genetic analyses across families and populations, we identified common genetic variation at the low-penetrance susceptibility HMMR locus (encoding for RHAMM) that modifies breast cancer risk among BRCA1, but probably not BRCA2, mutation carriers: n = 7,584, weighted hazard ratio ((w)HR) = 1.09 (95% CI 1.02-1.16), p(trend) = 0.017; and n = 3,965, (w)HR = 1.04 (95% CI 0.94-1.16), p(trend) = 0.43; respectively. Subsequently, studies of MCF10A apicobasal polarization revealed a central role for BRCA1 and RHAMM, together with AURKA and TPX2, in essential reorganization of microtubules. Mechanistically, reorganization is facilitated by BRCA1 and impaired by AURKA, which is regulated by negative feedback involving RHAMM and TPX2. Taken together, our data provide fundamental insight into apicobasal polarization through BRCA1 function, which may explain the expanded cell subsets and characteristic tumor type accompanying BRCA1 mutation, while also linking this process to sporadic breast cancer through perturbation of HMMR/RHAMM.


Subject(s)
BRCA1 Protein/metabolism , Breast Neoplasms/metabolism , Cell Polarity , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Aurora Kinase A , Aurora Kinases , BRCA1 Protein/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Breast/cytology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Polarity/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Genes, BRCA1 , Genes, BRCA2 , Genetic Predisposition to Disease , Genetic Variation , Genotype , HeLa Cells , Heterozygote , Humans , Microtubules/physiology , Microtubules/ultrastructure , Protein Serine-Threonine Kinases/metabolism , Receptors, Estrogen/analysis
18.
NPJ Microgravity ; 10(1): 49, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671027

ABSTRACT

One of the greatest challenges of humanity for deep space exploration is to fully understand how altered gravitational conditions affect human physiology. It is evident that the spaceflight environment causes multiple alterations to musculoskeletal, cardiovascular, immune and central nervous systems, to name a few known effects. To better characterize these biological effects, we compare gene expression datasets from microarray studies found in NASA GeneLab, part of the NASA Open Science Data Repository. In this review, we summarize these archived results for various tissues, emphasizing key genes which are highly reproducible in different mice or human experiments. Such exhaustive mining shows the potential of NASA Open Science data to identify and validate mechanisms taking place when mammalian organisms are exposed to microgravity or other spaceflight conditions. Our comparative meta-analysis findings highlight certain degrees of overlap and reproducibility in genes identified as differentially expressed within musculoskeletal tissues in each species across a variety of altered gravity conditions. However, the level of overlap between species was found to be significantly limited, partly attributed to the limited availability of human samples.

19.
Gigascience ; 132024 Jan 02.
Article in English | MEDLINE | ID: mdl-39283686

ABSTRACT

NASA's space life sciences research programs established a decades-long legacy of enhancing our ability to safely explore the cosmos. From Skylab and the Space Shuttle Program to the NASA Balloon Program and the International Space Station National Lab, these programs generated priceless data that continue to paint a vibrant picture of life in space. These data are available to the scientific community in various data repositories, including the NASA Ames Life Sciences Data Archive (ALSDA) and NASA GeneLab. Here we recognize the 30-year anniversary of data access through ALSDA and the 10-year anniversary of GeneLab.


Subject(s)
Biological Science Disciplines , Space Flight , United States National Aeronautics and Space Administration , United States , Databases, Factual , Humans
20.
Integr Biol (Camb) ; 162024 Jan 23.
Article in English | MEDLINE | ID: mdl-39299711

ABSTRACT

Cosmic radiation, composed of high charge and energy (HZE) particles, causes cellular DNA damage that can result in cell death or mutation that can evolve into cancer. In this work, a cell death model is applied to several cell lines exposed to HZE ions spanning a broad range of linear energy transfer (LET) values. We hypothesize that chromatin movement leads to the clustering of multiple double strand breaks (DSB) within one radiation-induced foci (RIF). The survival probability of a cell population is determined by averaging the survival probabilities of individual cells, which is function of the number of pairwise DSB interactions within RIF. The simulation code RITCARD was used to compute DSB. Two clustering approaches were applied to determine the number of RIF per cell. RITCARD outputs were combined with experimental data from four normal human cell lines to derive the model parameters and expand its predictions in response to ions with LET ranging from ~0.2 keV/µm to ~3000 keV/µm. Spherical and ellipsoidal nuclear shapes and two ion beam orientations were modeled to assess the impact of geometrical properties on cell death. The calculated average number of RIF per cell reproduces the saturation trend for high doses and high-LET values that is usually experimentally observed. The cell survival model generates the recognizable bell shape of LET dependence for the relative biological effectiveness (RBE). At low LET, smaller nuclei have lower survival due to increased DNA density and DSB clustering. At high LET, nuclei with a smaller irradiation area-either because of a smaller size or a change in beam orientation-have a higher survival rate due to a change in the distribution of DSB/RIF per cell. If confirmed experimentally, the geometric characteristics of cells would become a significant factor in predicting radiation-induced biological effects. Insight Box: High-charge and energy (HZE) ions are characterized by dense linear energy transfer (LET) that induce unique spatial distributions of DNA damage in cell nuclei that result in a greater biological effect than sparsely ionizing radiation like X-rays. HZE ions are a prominent component of galactic cosmic ray exposure during human spaceflight and specific ions are being used for radiotherapy. Here, we model DNA damage clustering at sub-micrometer scale to predict cell survival. The model is in good agreement with experimental data for a broad range of LET. Notably, the model indicates that nuclear geometry and ion beam orientation affect DNA damage clustering, which reveals their possible role in mediating cell radiosensitivity.


Subject(s)
Cell Death , Cosmic Radiation , DNA Breaks, Double-Stranded , Linear Energy Transfer , Humans , Cell Death/radiation effects , DNA Breaks, Double-Stranded/radiation effects , Cell Size/radiation effects , Cell Survival/radiation effects , Cell Line , Models, Biological , Computer Simulation , Dose-Response Relationship, Radiation , Cluster Analysis , Cell Nucleus/radiation effects , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/metabolism , Chromatin/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL