Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
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.

2.
Nat Commun ; 15(1): 4954, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862516

RESUMEN

Spaceflight induces an immune response in astronauts. To better characterize this effect, we generated single-cell, multi-ome, cell-free RNA (cfRNA), biochemical, and hematology data for the SpaceX Inspiration4 (I4) mission crew. We found that 18 cytokines/chemokines related to inflammation, aging, and muscle homeostasis changed after spaceflight. In I4 single-cell multi-omics data, we identified a "spaceflight signature" of gene expression characterized by enrichment in oxidative phosphorylation, UV response, immune function, and TCF21 pathways. We confirmed the presence of this signature in independent datasets, including the NASA Twins Study, the I4 skin spatial transcriptomics, and 817 NASA GeneLab mouse transcriptomes. Finally, we observed that (1) T cells showed an up-regulation of FOXP3, (2) MHC class I genes exhibited long-term suppression, and (3) infection-related immune pathways were associated with microbiome shifts. In summary, this study reveals conserved and distinct immune disruptions occurring and details a roadmap for potential countermeasures to preserve astronaut health.


Asunto(s)
Análisis de la Célula Individual , Vuelo Espacial , Transcriptoma , Animales , Femenino , Masculino , Humanos , Ratones , Astronautas , Citocinas/metabolismo , Linfocitos T/inmunología , Factores Sexuales , Perfilación de la Expresión Génica , Fosforilación Oxidativa
3.
Commun Biol ; 7(1): 692, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862620

RESUMEN

Organismal adaptations to spaceflight have been characterized at the molecular level in model organisms, including Drosophila and C. elegans. Here, we extend molecular work to energy metabolism and sex hormone signaling in mice and humans. We found spaceflight induced changes in insulin and estrogen signaling in rodents and humans. Murine changes were most prominent in the liver, where we observed inhibition of insulin and estrogen receptor signaling with concomitant hepatic insulin resistance and steatosis. Based on the metabolic demand, metabolic pathways mediated by insulin and estrogen vary among muscles, specifically between the soleus and extensor digitorum longus. In humans, spaceflight induced changes in insulin and estrogen related genes and pathways. Pathway analysis demonstrated spaceflight induced changes in insulin resistance, estrogen signaling, stress response, and viral infection. These data strongly suggest the need for further research on the metabolic and reproductive endocrinologic effects of space travel, if we are to become a successful interplanetary species.


Asunto(s)
Estrógenos , Insulina , Vuelo Espacial , Animales , Insulina/metabolismo , Estrógenos/metabolismo , Humanos , Ratones , Masculino , Femenino , Transcriptoma , Transducción de Señal , Ratones Endogámicos C57BL , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Hígado/metabolismo , Adulto , Regulación de la Expresión Génica
4.
Commun Med (Lond) ; 4(1): 106, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862781

RESUMEN

BACKGROUND: Spaceflight poses a unique set of challenges to humans and the hostile spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. METHODS: To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on space flown murine transcriptomic datasets focused on the skin, biochemical profiles of 50 NASA astronauts and human transcriptomic datasets generated from blood and hair samples of JAXA astronauts, as well as blood samples obtained from the NASA Twins Study, and skin and blood samples from the first civilian commercial mission, Inspiration4. RESULTS: Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation are identified as potential drivers for skin health risks during spaceflight. Additionally, a machine learning model is utilized to determine gene pairings associated with spaceflight response in the skin. While we identified spaceflight-induced dysregulation, such as alterations in genes associated with skin barrier function and collagen formation, our results also highlight the remarkable ability for organisms to re-adapt back to Earth via post-flight re-tuning of gene expression. CONCLUSION: Our findings can guide future research on developing countermeasures for mitigating spaceflight-associated skin damage.


Spaceflight is a hostile environment which can lead to health problems in astronauts, including in the skin. It is not currently well understood why these skin problems occur. Here, we analyzed data from the skin of space flown mice and astronauts to try and identify possible explanations for these skin problems. It appears that changes in the activation of genes related to damage to DNA, skin barrier health, and mitochondria (the energy-producing parts of cells) may play a role in these skin problems. Further research will be needed to confirm exactly how these changes influence skin health, which could lead to solutions for preventing and managing such issues in astronauts.

5.
Nat Microbiol ; 9(7): 1661-1675, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38862604

RESUMEN

Maintenance of astronaut health during spaceflight will require monitoring and potentially modulating their microbiomes. However, documenting microbial shifts during spaceflight has been difficult due to mission constraints that lead to limited sampling and profiling. Here we executed a six-month longitudinal study to quantify the high-resolution human microbiome response to three days in orbit for four individuals. Using paired metagenomics and metatranscriptomics alongside single-nuclei immune cell profiling, we characterized time-dependent, multikingdom microbiome changes across 750 samples and 10 body sites before, during and after spaceflight at eight timepoints. We found that most alterations were transient across body sites; for example, viruses increased in skin sites mostly during flight. However, longer-term shifts were observed in the oral microbiome, including increased plaque-associated bacteria (for example, Fusobacteriota), which correlated with immune cell gene expression. Further, microbial genes associated with phage activity, toxin-antitoxin systems and stress response were enriched across multiple body sites. In total, this study reveals in-depth characterization of microbiome and immune response shifts experienced by astronauts during short-term spaceflight and the associated changes to the living environment, which can help guide future missions, spacecraft design and space habitat planning.


Asunto(s)
Astronautas , Bacterias , Metagenómica , Microbiota , Vuelo Espacial , Humanos , Estudios Longitudinales , Microbiota/inmunología , Bacterias/clasificación , Bacterias/genética , Bacterias/inmunología , Masculino , Perfilación de la Expresión Génica , Adulto , Persona de Mediana Edad , Femenino , Transcriptoma , Multiómica
6.
Nat Commun ; 15(1): 4773, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862494

RESUMEN

Spaceflight can change metabolic, immunological, and biological homeostasis and cause skin rashes and irritation, yet the molecular basis remains unclear. To investigate the impact of short-duration spaceflight on the skin, we conducted skin biopsies on the Inspiration4 crew members before (L-44) and after (R + 1) flight. Leveraging multi-omics assays including GeoMx™ Digital Spatial Profiler, single-cell RNA/ATAC-seq, and metagenomics/metatranscriptomics, we assessed spatial gene expressions and associated microbial and immune changes across 95 skin regions in four compartments: outer epidermis, inner epidermis, outer dermis, and vasculature. Post-flight samples showed significant up-regulation of genes related to inflammation and KRAS signaling across all skin regions. These spaceflight-associated changes mapped to specific cellular responses, including altered interferon responses, DNA damage, epithelial barrier disruptions, T-cell migration, and hindered regeneration were located primarily in outer tissue compartments. We also linked epithelial disruption to microbial shifts in skin swab and immune cell activity to PBMC single-cell data from the same crew and timepoints. Our findings present the inaugural collection and examination of astronaut skin, offering insights for future space missions and response countermeasures.


Asunto(s)
Inflamación , Proteínas Proto-Oncogénicas p21(ras) , Piel , Vuelo Espacial , Humanos , Piel/inmunología , Piel/metabolismo , Piel/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Inflamación/inmunología , Inflamación/genética , Inflamación/metabolismo , Masculino , Análisis de la Célula Individual , Adulto , Persona de Mediana Edad , Femenino , Metagenómica/métodos , Perfilación de la Expresión Génica , Multiómica
7.
Nat Commun ; 15(1): 4825, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862542

RESUMEN

Our previous research revealed a key microRNA signature that is associated with spaceflight that can be used as a biomarker and to develop countermeasure treatments to mitigate the damage caused by space radiation. Here, we expand on this work to determine the biological factors rescued by the countermeasure treatment. We performed RNA-sequencing and transcriptomic analysis on 3D microvessel cell cultures exposed to simulated deep space radiation (0.5 Gy of Galactic Cosmic Radiation) with and without the antagonists to three microRNAs: miR-16-5p, miR-125b-5p, and let-7a-5p (i.e., antagomirs). Significant reduction of inflammation and DNA double strand breaks (DSBs) activity and rescue of mitochondria functions are observed after antagomir treatment. Using data from astronaut participants in the NASA Twin Study, Inspiration4, and JAXA missions, we reveal the genes and pathways implicated in the action of these antagomirs are altered in humans. Our findings indicate a countermeasure strategy that can potentially be utilized by astronauts in spaceflight missions to mitigate space radiation damage.


Asunto(s)
Astronautas , Radiación Cósmica , MicroARNs , Vuelo Espacial , MicroARNs/genética , MicroARNs/metabolismo , Humanos , Radiación Cósmica/efectos adversos , Roturas del ADN de Doble Cadena/efectos de la radiación , Traumatismos por Radiación/genética , Traumatismos por Radiación/prevención & control , Masculino , Mitocondrias/efectos de la radiación , Mitocondrias/metabolismo , Mitocondrias/genética , Femenino , Adulto
8.
Eur J Hum Genet ; 32(1): 10-20, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37938797

RESUMEN

COVID-19, the disease caused by SARS-CoV-2, has caused significant morbidity and mortality worldwide. The betacoronavirus continues to evolve with global health implications as we race to learn more to curb its transmission, evolution, and sequelae. The focus of this review, the second of a three-part series, is on the biological effects of the SARS-CoV-2 virus on post-acute disease in the context of tissue and organ adaptations and damage. We highlight the current knowledge and describe how virological, animal, and clinical studies have shed light on the mechanisms driving the varied clinical diagnoses and observations of COVID-19 patients. Moreover, we describe how investigations into SARS-CoV-2 effects have informed the understanding of viral pathogenesis and provide innovative pathways for future research on the mechanisms of viral diseases.


Asunto(s)
COVID-19 , Animales , Humanos , SARS-CoV-2
9.
NAR Genom Bioinform ; 5(4): lqad092, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37859800

RESUMEN

Given the current status of coronavirus disease 2019 (COVID-19) as a global pandemic, it is of high priority to gain a deeper understanding of the disease's development and how the virus impacts its host. Adenosine (A)-to-Inosine (I) RNA editing is a post-transcriptional modification, catalyzed by the ADAR family of enzymes, that can be considered part of the inherent cellular defense mechanism as it affects the innate immune response in a complex manner. It was previously reported that various viruses could interact with the host's ADAR enzymes, resulting in epigenetic changes both to the virus and the host. Here, we analyze RNA-seq of nasopharyngeal swab specimens as well as whole-blood samples of COVID-19 infected individuals and show a significant elevation in the global RNA editing activity in COVID-19 compared to healthy controls. We also detect specific coding sites that exhibit higher editing activity. We further show that the increment in editing activity during the disease is temporary and returns to baseline shortly after the symptomatic period. These significant epigenetic changes may contribute to the immune system response and affect adverse outcomes seen in post-viral cases.

10.
Res Sq ; 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37886447

RESUMEN

Maintenance of astronaut health during spaceflight will require monitoring and potentially modulating their microbiomes, which play a role in some space-derived health disorders. However, documenting the response of microbiota to spaceflight has been difficult thus far due to mission constraints that lead to limited sampling. Here, we executed a six-month longitudinal study centered on a three-day flight to quantify the high-resolution microbiome response to spaceflight. Via paired metagenomics and metatranscriptomics alongside single immune profiling, we resolved a microbiome "architecture" of spaceflight characterized by time-dependent and taxonomically divergent microbiome alterations across 750 samples and ten body sites. We observed pan-phyletic viral activation and signs of persistent changes that, in the oral microbiome, yielded plaque-associated pathobionts with strong associations to immune cell gene expression. Further, we found enrichments of microbial genes associated with antibiotic production, toxin-antitoxin systems, and stress response enriched universally across the body sites. We also used strain-level tracking to measure the potential propagation of microbial species from the crew members to each other and the environment, identifying microbes that were prone to seed the capsule surface and move between the crew. Finally, we identified associations between microbiome and host immune cell shifts, proposing both a microbiome axis of immune changes during flight as well as the sources of some of those changes. In summary, these datasets and methods reveal connections between crew immunology, the microbiome, and their likely drivers and lay the groundwork for future microbiome studies of spaceflight.

11.
Res Sq ; 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36798347

RESUMEN

Spaceflight poses a unique set of challenges to humans and the hostile Spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on spaceflown murine transcriptomic datasets focused on the skin, biomedical profiles from fifty NASA astronauts, and confirmation via transcriptomic data from JAXA astronauts, the NASA Twins Study, and the first civilian commercial mission, Inspiration4. Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation were determined to be involved with skin health risks during Spaceflight. Additionally, a machine learning model was utilized to determine key genes driving Spaceflight response in the skin. These results can be used for determining potential countermeasures to mitigate Spaceflight damage to the skin.

13.
J Hepatol ; 78(1): 45-56, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36049612

RESUMEN

BACKGROUND & AIMS: A number of genetic polymorphisms have been associated with susceptibility to or protection against non-alcoholic fatty liver disease (NAFLD), but the underlying mechanisms remain unknown. Here, we focused on the rs738409 C>G single nucleotide polymorphism (SNP), which produces the I148M variant of patatin-like phospholipase domain-containing protein 3 (PNPLA3) and is strongly associated with NAFLD. METHODS: To enable mechanistic dissection, we developed a human pluripotent stem cell (hPSC)-derived multicellular liver culture by incorporating hPSC-derived hepatocytes, hepatic stellate cells, and macrophages. We first applied this liver culture to model NAFLD by utilising a lipotoxic milieu reflecting the circulating levels of disease risk factors in affected individuals. We then created an isogenic pair of liver cultures differing only at rs738049 and compared NAFLD phenotype development. RESULTS: Our hPSC-derived liver culture recapitulated many key characteristics of NAFLD development and progression including lipid accumulation and oxidative stress, inflammatory response, and stellate cell activation. Under the lipotoxic conditions, the I148M variant caused the enhanced development of NAFLD phenotypes. These differences were associated with elevated IL-6/signal transducer and activator of transcription 3 (STAT3) activity in liver cultures, consistent with transcriptomic data of liver biopsies from individuals carrying the rs738409 SNP. Dampening IL-6/STAT3 activity alleviated the I148M-mediated susceptibility to NAFLD, whereas boosting it in wild-type liver cultures enhanced NAFLD development. Finally, we attributed this elevated IL-6/STAT3 activity in liver cultures carrying the rs738409 SNP to increased NF-κB activity. CONCLUSIONS: Our study thus reveals a potential causal link between elevated IL-6/STAT3 activity and 148M-mediated susceptibility to NAFLD. IMPACT AND IMPLICATIONS: An increasing number of genetic variants manifest in non-alcoholic fatty liver disease (NAFLD) development and progression; however, the underlying mechanisms remain elusive. To study these variants in human-relevant systems, we developed an induced pluripotent stem cell-derived multicellular liver culture and focused on a common genetic variant (i.e. rs738409 in PNPLA3). Our findings not only provide mechanistic insight, but also a potential therapeutic strategy for NAFLD driven by this genetic variant in PNPLA3. Our liver culture is therefore a useful platform for exploring genetic variants in NAFLD development.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Fosfolipasas A2 Calcio-Independiente , Humanos , Predisposición Genética a la Enfermedad , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Fosfolipasas A2 Calcio-Independiente/genética , Polimorfismo de Nucleótido Simple , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
14.
Foods ; 12(23)2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38231883

RESUMEN

With the increasing number of people affected by gluten consumption-related diseases, adhering to a gluten-free (GF) diet is the most effective preventive measure. Herein, we aimed to isolate and characterize the functional properties of autochthonous lactic acid bacteria (LAB) and yeast from various GF sourdoughs to determine their suitability in starter cultures for sourdough preparation. Three LAB, Weissella confusa BAQ2, Lactobacillus brevis AQ2, Leuconostoc citreum YC2, and Saccharomyces cerevisiae BW1, were identified. The isolated LAB exhibited greater TTA, faster acidification rates, and higher acid tolerance than commercial LAB. W. confusa BAQ2 exhibited the highest EPS production, W. confusa BAQ2 and L. brevis AQ2 showed high maltose utilization, and S. cerevisiae BW1 exhibited the highest CO2 production rate. Accordingly, all four microbial strains were mixed for the starter culture. The sourdough prepared with starter cultures exhibited differences in gas production depending on fermentation time, which influenced the volume of GF bread dough. GF bread prepared with fermented sourdough exhibited a 16% higher specific volume and enhanced crumb firmness and elasticity than that prepared using non-fermented sourdough. Thus, autochthonous LAB strains isolated from various GF sourdoughs can be used together to improve the quality of sourdough bread, demonstrating their potential for use in starter cultures for GF sourdough production.

15.
Front Vet Sci ; 10: 1280028, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38352169

RESUMEN

Introduction: Transarterial embolization (TAE) is one of the treatment options for liver masses that are not suitable for surgery and they have been applied in veterinary medicine for about 20 years, but surgical resection is considered as the first treatment option, and only a few case reports and articles about TAE in dogs have been published. Although understanding of vascular anatomy for the procedure is important, previous studies lack of the information about hepatic artery anatomy in small and toy-breed dogs. Due to the introduction of 3D print in veterinary medicine, it is now possible to make 3D models for preoperative planning. The purpose of this study is to understand the hepatic arterial vascular structure of various sizes and breeds of dogs, and to develop 3D-printed canine artery models with and without hepatic tumors to simulate TAE procedure. Methods: CT images of a total of 84 dogs with normal hepatic arteries were analyzed, and the mean value and standard deviation of body weight, celiac artery size, and hepatic artery size were 6.47 ± 4.44 kg, 3.28 ± 0.77 mm, and 2.14 ± 0.43 mm, respectively. Results: It was established that type 2-2-1, which has two separate hepatic branches-the right medial and left branch and the right lateral branch that runs to the right lateral lobe and caudate process-is the most prevalent of the hepatic artery branch types, as it was in the previous study. The review of 65 CT images of dogs with hepatic tumors showed that 44.6% (29/65) had multifocal lesions in multiple lobes, for which TAE can be recommended. Discussion: Based on the result, a 3D model of the normal canine hepatic artery and the hepatic tumor was made using one representative case from each group, and despite the models having some limitations in reflecting the exact tactile and velocity of blood vessels, TAE procedure was successfully simulated using both models.

16.
Genome Biol ; 23(1): 256, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36514162

RESUMEN

Spatial omics technologies enable a deeper understanding of cellular organizations and interactions within a tissue of interest. These assays can identify specific compartments or regions in a tissue with differential transcript or protein abundance, delineate their interactions, and complement other methods in defining cellular phenotypes. A variety of spatial methodologies are being developed and commercialized; however, these techniques differ in spatial resolution, multiplexing capability, scale/throughput, and coverage. Here, we review the current and prospective landscape of single cell to subcellular resolution spatial omics technologies and analysis tools to provide a comprehensive picture for both research and clinical applications.


Asunto(s)
Genómica , Proteómica , Genómica/métodos , Proteómica/métodos , Estudios Prospectivos
17.
Cell Rep Methods ; 2(11): 100325, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36452864

RESUMEN

Single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) have experienced rapid development in recent years. The findings of spaceflight-based scRNA-seq and SRT investigations are likely to improve our understanding of life in space and our comprehension of gene expression in various cell systems and tissue dynamics. However, compared to their Earth-based counterparts, gene expression experiments conducted in spaceflight have not experienced the same pace of development. Out of the hundreds of spaceflight gene expression datasets available, only a few used scRNA-seq and SRT. In this perspective piece, we explore the growing importance of scRNA-seq and SRT in space biology and discuss the challenges and considerations relevant to robust experimental design to enable growth of these methods in the field.


Asunto(s)
Vuelo Espacial , Transcriptoma , Transcriptoma/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica/métodos
18.
Nat Cell Biol ; 24(7): 1064-1076, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35787684

RESUMEN

The pancreas and liver arise from a common pool of progenitors. However, the underlying mechanisms that drive their lineage diversification from the foregut endoderm are not fully understood. To tackle this question, we undertook a multifactorial approach that integrated human pluripotent-stem-cell-guided differentiation, genome-scale CRISPR-Cas9 screening, single-cell analysis, genomics and proteomics. We discovered that HHEX, a transcription factor (TF) widely recognized as a key regulator of liver development, acts as a gatekeeper of pancreatic lineage specification. HHEX deletion impaired pancreatic commitment and unleashed an unexpected degree of cellular plasticity towards the liver and duodenum fates. Mechanistically, HHEX cooperates with the pioneer TFs FOXA1, FOXA2 and GATA4, shared by both pancreas and liver differentiation programmes, to promote pancreas commitment, and this cooperation restrains the shared TFs from activating alternative lineages. These findings provide a generalizable model for how gatekeeper TFs like HHEX orchestrate lineage commitment and plasticity restriction in broad developmental contexts.


Asunto(s)
Endodermo , Páncreas , Diferenciación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Páncreas/metabolismo , Factores de Transcripción
19.
Sci Rep ; 12(1): 9828, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35701572

RESUMEN

The aim of this study is to investigate the differences in polysomnographic and cephalometric features according to positional and rapid eye movement (REM) sleep dependencies in obstructive sleep apnea patients. Standard polysomnography and cephalometric analyses were performed on 133 OSA patients. The subjects were categorized into positional and non-positional, and REM-related and not-REM-related OSA groups according to positional and REM sleep dependency on severity of sleep apnea. Polysomnographic and cephalometric parameters were compared between groups. Positional and REM-related OSA patients showed significantly lower non-supine apnea-hypopnea index (AHI), non-REM (NREM) AHI and overall AHI and higher NREM oxygen saturation (SpO2) and mean SpO2 compared to non-positional and not-REM-related OSA patients, respectively. Cephalometric features between positional and non-positional OSA patients did not show any significant differences. However, REM-related OSA patients showed significantly larger inferior oral airway space and shorter perpendicular distance between mandibular plane and anterior hyoid bone and the distance between uvula and posterior nasal spine, and narrower maximum width of soft palate than not-REM-related OSA patients. Positional and REM-related OSA patients have lower severity of sleep apnea, suggesting the possibility of lower collapsibility of the upper airway. REM sleep dependency was associated with anatomical factors, while positional dependency did not show such a tendency.


Asunto(s)
Síndromes de la Apnea del Sueño , Apnea Obstructiva del Sueño , Cefalometría , Humanos , Polisomnografía , Sueño REM
20.
ISME J ; 16(6): 1636-1646, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35241788

RESUMEN

Prochlorococcus and Synechococcus are the most abundant photosynthesizing organisms in the oceans. Gene content variation among picocyanobacterial populations in separate ocean basins often mirrors the selective pressures imposed by the region's distinct biogeochemistry. By pairing genomic datasets with trace metal concentrations from across the global ocean, we show that the genomic capacity for siderophore-mediated iron uptake is widespread in Synechococcus and low-light adapted Prochlorococcus populations from deep chlorophyll maximum layers of iron-depleted regions of the oligotrophic Pacific and S. Atlantic oceans: Prochlorococcus siderophore consumers were absent in the N. Atlantic ocean (higher new iron flux) but constituted up to half of all Prochlorococcus genomes from metagenomes in the N. Pacific (lower new iron flux). Picocyanobacterial siderophore consumers, like many other bacteria with this trait, also lack siderophore biosynthesis genes indicating that they scavenge exogenous siderophores from seawater. Statistical modeling suggests that the capacity for siderophore uptake is endemic to remote ocean regions where atmospheric iron fluxes are the smallest, especially at deep chlorophyll maximum and primary nitrite maximum layers. We argue that abundant siderophore consumers at these two common oceanographic features could be a symptom of wider community iron stress, consistent with prior hypotheses. Our results provide a clear example of iron as a selective force driving the evolution of marine picocyanobacteria.


Asunto(s)
Prochlorococcus , Synechococcus , Clorofila , Hierro , Metagenoma , Océanos y Mares , Océano Pacífico , Filogenia , Prochlorococcus/genética , Agua de Mar/microbiología , Sideróforos , Synechococcus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...