Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Brain Behav Immun ; 113: 303-316, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37516387

RESUMO

Metabolomics, proteomics and DNA methylome assays, when done in tandem from the same blood sample and analyzed together, offer an opportunity to evaluate the molecular basis of post-traumatic stress disorder (PTSD) course and pathogenesis. We performed separate metabolomics, proteomics, and DNA methylome assays on blood samples from two well-characterized cohorts of 159 active duty male participants with relatively recent onset PTSD (<1.5 years) and 300 male veterans with chronic PTSD (>7 years). Analyses of the multi-omics datasets from these two independent cohorts were used to identify convergent and distinct molecular profiles that might constitute potential signatures of severity and progression of PTSD and its comorbid conditions. Molecular signatures indicative of homeostatic processes such as signaling and metabolic pathways involved in cellular remodeling, neurogenesis, molecular safeguards against oxidative stress, metabolism of polyunsaturated fatty acids, regulation of normal immune response, post-transcriptional regulation, cellular maintenance and markers of longevity were significantly activated in the active duty participants with recent PTSD. In contrast, we observed significantly altered multimodal molecular signatures associated with chronic inflammation, neurodegeneration, cardiovascular and metabolic disorders, and cellular attritions in the veterans with chronic PTSD. Activation status of signaling and metabolic pathways at the early and late timepoints of PTSD demonstrated the differential molecular changes related to homeostatic processes at its recent and multi-system syndromes at its chronic phase. Molecular alterations in the recent PTSD seem to indicate some sort of recalibration or compensatory response, possibly directed in mitigating the pathological trajectory of the disorder.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Veteranos , Humanos , Masculino , Transtornos de Estresse Pós-Traumáticos/genética , Transtornos de Estresse Pós-Traumáticos/metabolismo , Epigenômica , Proteômica , Metabolômica
2.
Mol Psychiatry ; 27(12): 5062-5069, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36131047

RESUMO

Posttraumatic stress disorder (PTSD) is a heritable (h2 = 24-71%) psychiatric illness. Copy number variation (CNV) is a form of rare genetic variation that has been implicated in the etiology of psychiatric disorders, but no large-scale investigation of CNV in PTSD has been performed. We present an association study of CNV burden and PTSD symptoms in a sample of 114,383 participants (13,036 cases and 101,347 controls) of European ancestry. CNVs were called using two calling algorithms and intersected to a consensus set. Quality control was performed to remove strong outlier samples. CNVs were examined for association with PTSD within each cohort using linear or logistic regression analysis adjusted for population structure and CNV quality metrics, then inverse variance weighted meta-analyzed across cohorts. We examined the genome-wide total span of CNVs, enrichment of CNVs within specified gene-sets, and CNVs overlapping individual genes and implicated neurodevelopmental regions. The total distance covered by deletions crossing over known neurodevelopmental CNV regions was significant (beta = 0.029, SE = 0.005, P = 6.3 × 10-8). The genome-wide neurodevelopmental CNV burden identified explains 0.034% of the variation in PTSD symptoms. The 15q11.2 BP1-BP2 microdeletion region was significantly associated with PTSD (beta = 0.0206, SE = 0.0056, P = 0.0002). No individual significant genes interrupted by CNV were identified. 22 gene pathways related to the function of the nervous system and brain were significant in pathway analysis (FDR q < 0.05), but these associations were not significant once NDD regions were removed. A larger sample size, better detection methods, and annotated resources of CNV are needed to explore this relationship further.


Assuntos
Variações do Número de Cópias de DNA , Transtornos de Estresse Pós-Traumáticos , Humanos , Transtornos de Estresse Pós-Traumáticos/genética , Genoma , Encéfalo , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Predisposição Genética para Doença
3.
Curr Issues Mol Biol ; 44(8): 3711-3734, 2022 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-36005150

RESUMO

Countermeasures for radiation diagnosis, prognosis, and treatment are trailing behind the proliferation of nuclear energy and weaponry. Radiation injury mechanisms at the systems biology level are not fully understood. Here, mice skin biopsies at h2, d4, d7, d21, and d28 after exposure to 1, 3, 6, or 20 Gy whole-body ionizing radiation were evaluated for the potential application of transcriptional alterations in radiation diagnosis and prognosis. Exposure to 20 Gy was lethal by d7, while mice who received 1, 3, or 6 Gy survived the 28-day time course. A Sammon plot separated samples based on survival and time points (TPs) within lethal (20 Gy) and sublethal doses. The differences in the numbers, regulation mode, and fold change of significantly differentially transcribed genes (SDTGs, p < 0.05 and FC > 2) were identified between lethal and sublethal doses, and down and upregulation dominated transcriptomes during the first post-exposure week, respectively. The numbers of SDTGs and the percentages of upregulated ones revealed stationary downregulation post-lethal dose in contrast to responses to sublethal doses which were dynamic and largely upregulated. Longitudinal up/downregulated SDTGs ratios suggested delayed and extended responses with increasing IR doses in the sublethal range and lethal-like responses in late TPs. This was supported by the distributions of common and unique genes across TPs within each dose. Several genes with potential dosimetric marker applications were identified. Immune, fibrosis, detoxification, hematological, neurological, gastric, cell survival, migration, and proliferation radiation response pathways were identified, with the majority predicted to be activated after sublethal and inactivated after lethal exposures, particularly during the first post-exposure week.

4.
Mol Psychiatry ; 26(8): 4300-4314, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33339956

RESUMO

Post-traumatic stress disorder (PTSD) is a heterogeneous condition evidenced by the absence of objective physiological measurements applicable to all who meet the criteria for the disorder as well as divergent responses to treatments. This study capitalized on biological diversity observed within the PTSD group observed following epigenome-wide analysis of a well-characterized Discovery cohort (N = 166) consisting of 83 male combat exposed veterans with PTSD, and 83 combat veterans without PTSD in order to identify patterns that might distinguish subtypes. Computational analysis of DNA methylation (DNAm) profiles identified two PTSD biotypes within the PTSD+ group, G1 and G2, associated with 34 clinical features that are associated with PTSD and PTSD comorbidities. The G2 biotype was associated with an increased PTSD risk and had higher polygenic risk scores and a greater methylation compared to the G1 biotype and healthy controls. The findings were validated at a 3-year follow-up (N = 59) of the same individuals as well as in two independent, veteran cohorts (N = 54 and N = 38), and an active duty cohort (N = 133). In some cases, for example Dopamine-PKA-CREB and GABA-PKC-CREB signaling pathways, the biotypes were oppositely dysregulated, suggesting that the biotypes were not simply a function of a dimensional relationship with symptom severity, but may represent distinct biological risk profiles underpinning PTSD. The identification of two novel distinct epigenetic biotypes for PTSD may have future utility in understanding biological and clinical heterogeneity in PTSD and potential applications in risk assessment for active duty military personnel under non-clinician-administered settings, and improvement of PTSD diagnostic markers.


Assuntos
Militares , Transtornos de Estresse Pós-Traumáticos , Veteranos , Epigênese Genética/genética , Epigenoma , Humanos , Masculino , Transtornos de Estresse Pós-Traumáticos/genética
5.
Mol Psychiatry ; 26(9): 5011-5022, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32488126

RESUMO

Active-duty Army personnel can be exposed to traumatic warzone events and are at increased risk for developing post-traumatic stress disorder (PTSD) compared with the general population. PTSD is associated with high individual and societal costs, but identification of predictive markers to determine deployment readiness and risk mitigation strategies is not well understood. This prospective longitudinal naturalistic cohort study-the Fort Campbell Cohort study-examined the value of using a large multidimensional dataset collected from soldiers prior to deployment to Afghanistan for predicting post-deployment PTSD status. The dataset consisted of polygenic, epigenetic, metabolomic, endocrine, inflammatory and routine clinical lab markers, computerized neurocognitive testing, and symptom self-reports. The analysis was computed on active-duty Army personnel (N = 473) of the 101st Airborne at Fort Campbell, Kentucky. Machine-learning models predicted provisional PTSD diagnosis 90-180 days post deployment (random forest: AUC = 0.78, 95% CI = 0.67-0.89, sensitivity = 0.78, specificity = 0.71; SVM: AUC = 0.88, 95% CI = 0.78-0.98, sensitivity = 0.89, specificity = 0.79) and longitudinal PTSD symptom trajectories identified with latent growth mixture modeling (random forest: AUC = 0.85, 95% CI = 0.75-0.96, sensitivity = 0.88, specificity = 0.69; SVM: AUC = 0.87, 95% CI = 0.79-0.96, sensitivity = 0.80, specificity = 0.85). Among the highest-ranked predictive features were pre-deployment sleep quality, anxiety, depression, sustained attention, and cognitive flexibility. Blood-based biomarkers including metabolites, epigenomic, immune, inflammatory, and liver function markers complemented the most important predictors. The clinical prediction of post-deployment symptom trajectories and provisional PTSD diagnosis based on pre-deployment data achieved high discriminatory power. The predictive models may be used to determine deployment readiness and to determine novel pre-deployment interventions to mitigate the risk for deployment-related PTSD.


Assuntos
Militares , Transtornos de Estresse Pós-Traumáticos , Afeganistão , Estudos de Coortes , Humanos , Aprendizado de Máquina , Estudos Prospectivos , Fatores de Risco , Qualidade do Sono
6.
Mol Psychiatry ; 26(9): 4999-5009, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32382136

RESUMO

DNA methylation patterns at specific cytosine-phosphate-guanine (CpG) sites predictably change with age and can be used to derive "epigenetic age", an indicator of biological age, as opposed to merely chronological age. A relatively new estimator, called "DNAm GrimAge", is notable for its superior predictive ability in older populations regarding numerous age-related metrics like time-to-death, time-to-coronary heart disease, and time-to-cancer. PTSD is associated with premature mortality and frequently has comorbid physical illnesses suggestive of accelerated biological aging. This is the first study to assess DNAm GrimAge in PTSD patients. We investigated the acceleration of GrimAge relative to chronological age, denoted "AgeAccelGrim" in combat trauma-exposed male veterans with and without PTSD using cross-sectional and longitudinal data from two independent well-characterized veteran cohorts. In both cohorts, AgeAccelGrim was significantly higher in the PTSD group compared to the control group (N = 162, 1.26 vs -0.57, p = 0.001 and N = 53, 0.93 vs -1.60 Years, p = 0.008), suggesting accelerated biological aging in both cohorts with PTSD. In 3-year follow-up study of individuals initially diagnosed with PTSD (N = 26), changes in PTSD symptom severity were correlated with AgeAccelGrim changes (r = 0.39, p = 0.049). In addition, the loss of CD28 cell surface markers on CD8 + T cells, an indicator of T-cell senescence/exhaustion that is associated with biological aging, was positively correlated with AgeAccelGrim, suggesting an immunological contribution to the accelerated biological aging. Overall, our findings delineate cellular correlates of biological aging in combat-related PTSD, which may help explain the increased medical morbidity and mortality seen in this disease.


Assuntos
Metilação de DNA , Transtornos de Estresse Pós-Traumáticos , Idoso , Envelhecimento/genética , Estudos Transversais , Metilação de DNA/genética , Epigênese Genética , Epigenômica , Seguimentos , Humanos , Masculino , Transtornos de Estresse Pós-Traumáticos/genética
7.
Int J Mol Sci ; 23(20)2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36293361

RESUMO

Post-traumatic stress disorder (PTSD) is a highly debilitating psychiatric disorder that can be triggered by exposure to extreme trauma. Even if PTSD is primarily a psychiatric condition, it is also characterized by adverse somatic comorbidities. One illness commonly co-occurring with PTSD is Metabolic syndrome (MetS), which is defined by a set of health risk/resilience factors including obesity, elevated blood pressure, lower high-density lipoprotein cholesterol, higher low-density lipoprotein cholesterol, higher triglycerides, higher fasting blood glucose and insulin resistance. Here, phenotypic association between PTSD and components of MetS are tested on a military veteran cohort comprising chronic PTSD presentation (n = 310, 47% cases, 83% male). Consistent with previous observations, we found significant phenotypic correlation between the various components of MetS and PTSD severity scores. To examine if this observed symptom correlations stem from a shared genetic background, we conducted genetic correlation analysis using summary statistics data from large-scale genetic studies. Our results show robust positive genetic correlation between PTSD and MetS (rg[SE] = 0.33 [0.056], p = 4.74E-09), and obesity-related components of MetS (rg = 0.25, SE = 0.05, p = 6.4E-08). Prioritizing genomic regions with larger local genetic correlation implicate three significant loci. Overall, these findings show significant genetic overlap between PTSD and MetS, which may in part account for the markedly increased occurrence of MetS among PTSD patients.


Assuntos
Síndrome Metabólica , Transtornos de Estresse Pós-Traumáticos , Humanos , Masculino , Feminino , Transtornos de Estresse Pós-Traumáticos/complicações , Transtornos de Estresse Pós-Traumáticos/epidemiologia , Transtornos de Estresse Pós-Traumáticos/genética , Síndrome Metabólica/complicações , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/genética , Prevalência , Glicemia , Obesidade , Lipoproteínas HDL , Lipoproteínas LDL , Triglicerídeos , Colesterol
8.
Brain Behav Immun ; 91: 429-436, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33152445

RESUMO

Posttraumatic stress disorder (PTSD) is characterized by intrusive thoughts, avoidance, negative alterations in cognitions and mood, and arousal symptoms that adversely affect mental and physical health. Recent evidence links changes in DNA methylation of CpG cites to PTSD. Since clusters of proximal CpGs share similar methylation signatures, identification of PTSD-associated differentially methylated regions (DMRs) may elucidate the pathways defining differential risk and resilience of PTSD. Here we aimed to identify epigenetic differences associated with PTSD. DNA methylation data profiled from blood samples using the MethylationEPIC BeadChip were used to perform a DMR analysis in 187 PTSD cases and 367 trauma-exposed controls from the Grady Trauma Project (GTP). DMRs were assessed with R package bumphunter. We identified two regions that associate with PTSD after multiple test correction. These regions were in the gene body of HLA-DPB1 and in the promoter of SPATC1L. The DMR in HLA-DPB1 was associated with PTSD in an independent cohort. Both DMRs included CpGs whose methylation associated with nearby sequence variation (meQTL) and that associated with expression of their respective genes (eQTM). This study supports an emerging literature linking PTSD risk to genetic and epigenetic variation in the HLA region.


Assuntos
Proteínas do Citoesqueleto/genética , Metilação de DNA , Cadeias beta de HLA-DP/genética , Transtornos de Estresse Pós-Traumáticos , Epigênese Genética , Epigenômica , Humanos , Transtornos de Estresse Pós-Traumáticos/genética
9.
Mol Psychiatry ; 25(12): 3337-3349, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31501510

RESUMO

Post-traumatic stress disorder (PTSD) impacts many veterans and active duty soldiers, but diagnosis can be problematic due to biases in self-disclosure of symptoms, stigma within military populations, and limitations identifying those at risk. Prior studies suggest that PTSD may be a systemic illness, affecting not just the brain, but the entire body. Therefore, disease signals likely span multiple biological domains, including genes, proteins, cells, tissues, and organism-level physiological changes. Identification of these signals could aid in diagnostics, treatment decision-making, and risk evaluation. In the search for PTSD diagnostic biomarkers, we ascertained over one million molecular, cellular, physiological, and clinical features from three cohorts of male veterans. In a discovery cohort of 83 warzone-related PTSD cases and 82 warzone-exposed controls, we identified a set of 343 candidate biomarkers. These candidate biomarkers were selected from an integrated approach using (1) data-driven methods, including Support Vector Machine with Recursive Feature Elimination and other standard or published methodologies, and (2) hypothesis-driven approaches, using previous genetic studies for polygenic risk, or other PTSD-related literature. After reassessment of ~30% of these participants, we refined this set of markers from 343 to 28, based on their performance and ability to track changes in phenotype over time. The final diagnostic panel of 28 features was validated in an independent cohort (26 cases, 26 controls) with good performance (AUC = 0.80, 81% accuracy, 85% sensitivity, and 77% specificity). The identification and validation of this diverse diagnostic panel represents a powerful and novel approach to improve accuracy and reduce bias in diagnosing combat-related PTSD.


Assuntos
Militares , Transtornos de Estresse Pós-Traumáticos , Veteranos , Biomarcadores , Encéfalo , Humanos , Masculino , Transtornos de Estresse Pós-Traumáticos/diagnóstico , Transtornos de Estresse Pós-Traumáticos/genética
10.
Environ Res ; 192: 110245, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32987006

RESUMO

Natural communities of microbes inhabiting amphibian skin, the skin microbiome, are critical to supporting amphibian health and disease resistance. To enable the pro-active health assessment and management of amphibians on Army installations and beyond, we investigated the effects of acute (96h) munitions exposures to Rana pipiens (leopard frog) tadpoles and the associated skin microbiome, integrated with RNAseq-based transcriptomic responses in the tadpole host. Tadpoles were exposed to the legacy munition 2,4,6-trinitrotoluene (TNT), the new insensitive munition (IM) formulation, IMX-101, and the IM constituents nitroguinidine (NQ) and 1-methyl-3-nitroguanidine (MeNQ). The 96h LC50 values and 95% confidence intervals were 2.6 (2.4, 2.8) for ΣTNT and 68.2 (62.9, 73.9) for IMX-101, respectively. The NQ and MeNQ exposures caused no significant impacts on survival in 96h exposures even at maximum exposure levels of 3560 and 5285 mg/L, respectively. However, NQ and MeNQ, as well as TNT and IMX-101 exposures, all elicited changes in the tadpole skin microbiome profile, as evidenced by significantly increased relative proportions of the Proteobacteria with increasing exposure concentrations, and significantly decreased alpha-diversity in the NQ exposure. The potential for direct effects of munitions exposure on the skin microbiome were observed including increased abundance of munitions-tolerant phylogenetic groups, in addition to possible indirect effects on microbial flora where transcriptional responses suggestive of changes in skin mucus-layer properties, antimicrobial peptide production, and innate immune factors were observed in the tadpole host. Additional insights into the tadpole host's transcriptional response to munitions exposures indicated that TNT and IMX-101 exposures significantly enriched transcriptional expression within type-I and type-II xenobiotic metabolism pathways, where dose-responsive increases in expression were observed. Significant enrichment and increased transcriptional expression of heme and iron binding functions in the TNT exposures served as likely indicators of known mechanisms of TNT toxicity including hemolytic anemia and methemoglobinemia. The significant enrichment and dose-responsive decrease in transcriptional expression of cell cycle pathways in the IMX-101 exposures was consistent with previous observations in fish, while significant enrichment of immune-related function in response to NQ exposure were consistent with potential immune suppression at the highest NQ exposure concentration. Finally, the MeNQ exposures elicited significantly decreased transcriptional expression of keratin 16, type I, a gene likely involved in keratinization processes in amphibian skin. Overall, munitions showed the potential to alter tadpole skin microbiome composition and affect transcriptional profiles in the amphibian host, some suggestive of potential impacts on host health and immune status relevant to disease susceptibility.


Assuntos
Genômica , Microbiota , Animais , Larva , Filogenia , Rana pipiens
11.
Int J Mol Sci ; 22(6)2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33807089

RESUMO

Clinical, epidemiological, and experimental evidence demonstrate non-cancer, cardiovascular, and endocrine effects of ionizing radiation exposure including growth hormone deficiency, obesity, metabolic syndrome, diabetes, and hyperinsulinemia. Insulin-like growth factor-1 (IGF-1) signaling perturbations are implicated in development of cardiovascular disease and metabolic syndrome. The minipig is an emerging model for studying radiation effects given its high analogy to human anatomy and physiology. Here we use a minipig model to study late health effects of radiation by exposing male Göttingen minipigs to 1.9-2.0 Gy X-rays (lower limb tibias spared). Animals were monitored for 120 days following irradiation and blood counts, body weight, heart rate, clinical chemistry parameters, and circulating biomarkers were assessed longitudinally. Collagen deposition, histolopathology, IGF-1 signaling, and mRNA sequencing were evaluated in tissues. Our findings indicate a single exposure induced histopathological changes, attenuated circulating IGF-1, and disrupted cardiac IGF-1 signaling. Electrolytes, lipid profiles, liver and kidney markers, and heart rate and rhythm were also affected. In the heart, collagen deposition was significantly increased and transforming growth factor beta-1 (TGF-beta-1) was induced following irradiation; collagen deposition and fibrosis were also observed in the kidney of irradiated animals. Our findings show Göttingen minipigs are a suitable large animal model to study long-term effects of radiation exposure and radiation-induced inhibition of IGF-1 signaling may play a role in development of late organ injuries.


Assuntos
Biomarcadores , Fator de Crescimento Insulin-Like I/metabolismo , Miocárdio/metabolismo , Lesões por Radiação/metabolismo , Transdução de Sinais/efeitos da radiação , Animais , Células Sanguíneas/metabolismo , Células Sanguíneas/efeitos da radiação , Peso Corporal/efeitos da radiação , Colágeno/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Fibrose/etiologia , Regulação da Expressão Gênica/efeitos da radiação , Frequência Cardíaca/efeitos da radiação , Hematopoese/efeitos da radiação , Metabolismo dos Lipídeos/efeitos da radiação , Especificidade de Órgãos/efeitos da radiação , Lesões por Radiação/genética , Suínos
12.
Brain Behav Immun ; 80: 725-741, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31100372

RESUMO

BACKGROUND: Severe stress can have drastic and systemic effects with dire implications on the health and wellbeing of exposed individuals. Particularly, the effect of stress on the immune response to infection is of interest to public health because of its implications for vaccine efficacy and treatment strategies during stressful scenarios. Severe stress has previously been shown to cause an anergic state in the immune system that persists following exposure to a potent mitogen. METHODS: Transcriptome and microRNA changes were characterized using blood samples collected from U.S. Army Ranger candidates immediately before and after training, followed by exposure to representative pathogenic agents: Yersinia pestis, dengue virus 2, and Staphylococcal enterotoxin B (SEB). We employed experimental and computational approaches to characterize altered gene expression, processes, pathways, and regulatory networks mediating the host's response towards severe stress; to assess the protective immunity status of the stressed host towards infection; and to identify pathogen-induced biomarkers under severe stress conditions. RESULTS: We observed predicted inhibition of pathways significantly associated with lymphopoiesis, wound healing, inflammatory response, lymphocyte activation, apoptosis, and predicted activation of oxidative stress. Using weighted correlation network analyses, we demonstrated preservation of these pathways across infection and stress combinations. Regulatory networks comprising a common set of upstream regulators: transcription factors, microRNAs and post-translational regulators (kinases and phosphatases) may be drivers of molecular alterations leading to compromised protective immunity. Other sets of transcripts were persistently altered in both the pre- and post-stress conditions due to the host's response to each pathogenic agent, forming specific molecular signatures with the potential to distinguish infection from that of severe stress. CONCLUSIONS: Our results suggest that severe stress alters molecules implicated in the development of leukopoietic stem cells, thereby leading to depletion of cellular and molecular repertoires of protective immunity. Suppressed molecules mediating membrane trafficking of recycling endosomes, membrane translocation and localization of the antigen processing mechanisms and cell adhesions indicate suboptimal antigen presentation, impaired formation of productive immunological synapses, and inhibited T-cell activations. These factors may collectively be responsible for compromised protective immunity (infection susceptibility, delayed wound healing, and poor vaccine response) observed in people under severe stress.


Assuntos
Imunidade Inata/genética , Imunidade Inata/imunologia , Estresse Fisiológico/genética , Estresse Fisiológico/imunologia , Adulto , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Vírus da Dengue/imunologia , Enterotoxinas/imunologia , Humanos , Ativação Linfocitária/imunologia , Masculino , MicroRNAs/sangue , MicroRNAs/genética , Transcriptoma/genética , Yersinia pestis/imunologia
13.
J Neurosci Res ; 96(7): 1311-1323, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29633335

RESUMO

The bidirectional role of gut-brain axis that integrates the gut and central nervous system activities has recently been investigated. We studied "cage-within-cage resident-intruder" all-male model, where subject male mice (C57BL/6J) are exposed to aggressor mice (SJL albino), and gut microbiota-derived metabolites were identified in plasma after 10 days of exposure. We assessed 16S ribosomal RNA gene from fecal samples collected daily from these mice during the 10-day study. Alpha diversity using Chao indices indicated no change in diversity in aggressor-exposed samples. The abundance profile showed the top phyla were Firmicutes and Bacteroidetes, Tenericutes, Verrucomicrobia, Actinobacteria and Proteobacteria, respectively. The phyla Firmicutes and Bacteroidetes are vulnerable to PTSD-eliciting stress and the Firmicutes/Bacteroidetes ratio increases with stress. Principal coordinate analysis showed the control and aggressor-exposed samples cluster separately where samples from early time points (day 1-3) clustered together and were distinct from late time points (day 4-9). The genus-based analysis revealed all control time points clustered together and aggressor-exposed samples had multiple clusters. The decrease in proportion of Firmicutes after aggressor exposure persisted throughout the study. The proportion of Verrucomicrobia immediately decreased and was significantly shifted at most of the later time points. The genus Oscillospira, Lactobacillus, Akkermansia and Anaeroplasma are the top four genera that differed between control and stressor-exposed mice. The data showed immediate effect on microbiome composition during a 10 day time period of stress exposure. Studying the longitudinal effects of a stressor is an important step toward an improved mechanistic understanding of the microbiome dynamics.


Assuntos
Fezes/microbiologia , Microbioma Gastrointestinal , Transtornos de Estresse Pós-Traumáticos/microbiologia , Animais , Bacteroidetes/isolamento & purificação , Firmicutes/isolamento & purificação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Proteobactérias/isolamento & purificação
14.
Appl Environ Microbiol ; 84(21)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30217846

RESUMO

The experimental pathophysiology of organophosphorus (OP) chemical exposure has been extensively reported. Here, we describe an altered fecal bacterial biota and urine metabolome following intoxication with soman, a lipophilic G class chemical warfare nerve agent. Nonanesthetized Sprague-Dawley male rats were subcutaneously administered soman at 0.8 (subseizurogenic) or 1.0 (seizurogenic) of the 50% lethal dose (LD50) and evaluated for signs of toxicity. Animals were stratified based on seizing activity to evaluate effects of soman exposure on fecal bacterial biota and urine metabolites. Soman exposure reshaped fecal bacterial biota by altering Facklamia, Rhizobium, Bilophila, Enterobacter, and Morganella genera of the Firmicutes and Proteobacteria phyla, some of which are known to hydrolyze OP chemicals. However, analogous changes were not observed in the bacterial biota of the ileum, which remained the same irrespective of dose or seizing status of animals after soman intoxication. However, at 75 days after soman exposure, the bacterial biota stabilized and no differences were observed between groups. Interestingly, in considering just the seizing status of animals, we found that the urine metabolomes were markedly different. Leukotriene C4, kynurenic acid, 5-hydroxyindoleacetic acid, norepinephrine, and aldosterone were excreted at much higher rates at 72 h in seizing animals, consistent with early multiorgan involvement during soman poisoning. These findings demonstrate the feasibility of using the dysbiosis of fecal bacterial biota in combination with urine metabolome alterations as forensic evidence for presymptomatic OP exposure temporally to enable administration of neuroprotective therapies of the future.IMPORTANCE The paucity of assays to determine physiologically relevant OP exposure presents an opportunity to explore the use of fecal bacteria as sentinels in combination with urine to assess changes in the exposed host. Recent advances in sequencing technologies and computational approaches have enabled researchers to survey large community-level changes of gut bacterial biota and metabolomic changes in various biospecimens. Here, we profiled changes in fecal bacterial biota and urine metabolome following a chemical warfare nerve agent exposure. The significance of this work is a proof of concept that the fecal bacterial biota and urine metabolites are two separate biospecimens rich in surrogate indicators suitable for monitoring OP exposure. The larger value of such an approach is that assays developed on the basis of these observations can be deployed in any setting with moderate clinical chemistry and microbiology capability. This can enable estimation of the affected radius as well as screening, triage, or ruling out of suspected cases of exposures in mass casualty scenarios, transportation accidents involving hazardous materials, refugee movements, humanitarian missions, and training settings when coupled to an established and validated decision tree with clinical features.


Assuntos
Bactérias/efeitos dos fármacos , Biota/efeitos dos fármacos , Fezes/microbiologia , Agentes Neurotóxicos/intoxicação , Convulsões/metabolismo , Soman/intoxicação , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/etiologia , Convulsões/microbiologia , Convulsões/urina , Soman/administração & dosagem , Urina/química
15.
Metabolomics ; 15(1): 2, 2018 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-30830480

RESUMO

INTRODUCTION: Pneumonic plague is caused by the aerosolized form of Yersinia pestis and is a highly virulent infection with complex clinical consequences, and without treatment, the fatality rate approaches 100%. The exact mechanisms of disease progression are unclear, with limited work done using metabolite profiling to study disease progression. OBJECTIVE: The aim of this pilot study was to profile the plasma metabolomics in an animal model of Y. pestis infection. METHODS: In this study, African Green monkeys were challenged with the highly virulent, aerosolized Y. pestis strain CO92, and untargeted metabolomics profiling of plasma was performed using liquid and gas chromatography with mass spectrometry. RESULTS: At early time points post-exposure, we found significant increases in polyunsaturated, long chain fatty acid metabolites with p values ranging from as low as 0.000001 (ratio = 1.94) for the metabolite eicosapentaenoate to 0.04 (ratio = 1.36) for the metabolite adrenate when compared to time-matched controls. Multiple acyl carnitines metabolites were increased at earlier time points and could be a result of fatty acid oxidation defects with p values ranging from as low as 0.00001 (ratio = 2.95) for the metabolite octanoylcarnitine to 0.04 (ratio = 1.33) for metabolite deoxycarnitine when compared to time-matched controls. Dicarboxylic acids are important metabolic products of fatty acids oxidation, and when compared to time matched controls, were higher at earlier time points where metabolite tetradecanedioate has a ratio of 4.09 with significant p value of 0.000002 and adipate with a ratio of 1.12 and p value of 0.004. The metabolites from lysolipids (with significant p values ranging from 0.00006 for 1-oleoylglycerophosphoethanolamine to 0.04 for 1-stearoylglycerophosphoethanolamine and a ratio of 0.47 and 0.78, respectively) and bile acid metabolism (with significant p values ranging from 0.02 for cholate to 0.04 for deoxycholate and a ratio of 0.39 and 0.66, respectively) pathways were significantly lower compared to their time-matched controls during the entire course of infection. Metabolite levels from amino acid pathways were disrupted, and a few from the leucine, isoleucine and valine pathway were significantly higher (p values ranging from 0.002 to 0.04 and ratios ranging from 1.3 to 1.5, respectively), whereas metabolites from the urea cycle, arginine and proline pathways were significantly lower (p values ranging from 0.00008 to 0.02 and ratios ranging from 0.5 to 0.7, respectively) during the course of infection. CONCLUSIONS: The involvement of several lipid pathways post-infection suggested activation of pathways linked to inflammation and oxidative stress. Metabolite data further showed increased energy demand, and multiple metabolites indicated potential hepatic dysfunction. Integration of blood metabolomics and transcriptomics data identified linoleate as a core metabolite with cross-talk with multiple genes from various time points. Collectively, the data from this study provided new insights into the mechanisms of Y. pestis pathogenesis that may aid in development of therapeutics.


Assuntos
Metabolômica/métodos , Yersinia pestis/metabolismo , Animais , Betaína/análogos & derivados , Betaína/metabolismo , Carnitina/metabolismo , Chlorocebus aethiops , Modelos Animais de Doenças , Cromatografia Gasosa-Espectrometria de Massas
16.
Am J Physiol Gastrointest Liver Physiol ; 312(6): G559-G571, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28336545

RESUMO

The magnitude, temporal dynamics, and physiological effects of intestinal microbiome responses to physiological stress are poorly characterized. This study used a systems biology approach and a multiple-stressor military training environment to determine the effects of physiological stress on intestinal microbiota composition and metabolic activity, as well as intestinal permeability (IP). Soldiers (n = 73) were provided three rations per day with or without protein- or carbohydrate-based supplements during a 4-day cross-country ski-march (STRESS). IP was measured before and during STRESS. Blood and stool samples were collected before and after STRESS to measure inflammation, stool microbiota, and stool and plasma global metabolite profiles. IP increased 62 ± 57% (mean ± SD, P < 0.001) during STRESS independent of diet group and was associated with increased inflammation. Intestinal microbiota responses were characterized by increased α-diversity and changes in the relative abundance of >50% of identified genera, including increased abundance of less dominant taxa at the expense of more dominant taxa such as Bacteroides Changes in intestinal microbiota composition were linked to 23% of metabolites that were significantly altered in stool after STRESS. Together, pre-STRESS Actinobacteria relative abundance and changes in serum IL-6 and stool cysteine concentrations accounted for 84% of the variability in the change in IP. Findings demonstrate that a multiple-stressor military training environment induced increases in IP that were associated with alterations in markers of inflammation and with intestinal microbiota composition and metabolism. Associations between IP, the pre-STRESS microbiota, and microbiota metabolites suggest that targeting the intestinal microbiota could provide novel strategies for preserving IP during physiological stress.NEW & NOTEWORTHY Military training, a unique model for studying temporal dynamics of intestinal barrier and intestinal microbiota responses to stress, resulted in increased intestinal permeability concomitant with changes in intestinal microbiota composition and metabolism. Prestress intestinal microbiota composition and changes in fecal concentrations of metabolites linked to the microbiota were associated with increased intestinal permeability. Findings suggest that targeting the intestinal microbiota could provide novel strategies for mitigating increases in intestinal permeability during stress.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal , Absorção Intestinal , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Estresse Fisiológico , Adolescente , Fatores Etários , Carboidratos da Dieta/administração & dosagem , Carboidratos da Dieta/metabolismo , Proteínas Alimentares/administração & dosagem , Proteínas Alimentares/metabolismo , Metabolismo Energético , Fezes/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/sangue , Masculino , Metabolômica/métodos , Medicina Militar , Noruega , Estado Nutricional , Permeabilidade , Resistência Física , Biologia de Sistemas , Fatores de Tempo , Adulto Jovem
18.
Proc Natl Acad Sci U S A ; 111(8): 3188-93, 2014 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-24516145

RESUMO

Posttraumatic stress disorder (PTSD) is a common condition induced by life-threatening stress, such as that experienced by soldiers under battlefield conditions. Other than the commonly recognized behavioral and psychological dysfunction, epidemiological studies have also revealed that PTSD patients have a higher risk of other diseases, such as cardiovascular disorders. Using a PTSD mouse model, we investigated the longitudinal transcriptomic changes in heart tissues after the exposure to stress through intimidation. Our results revealed acute heart injury associated with the traumatic experience, reflecting the underlying biological injury processes of the immune response, extracellular matrix remodeling, epithelial-to-mesenchymal cell transitions, and cell proliferation. Whether this type of injury has any long-term effects on heart function is yet to be determined. The differing responses to stress leading to acute heart injury in different inbred strains of mice also suggest that this response has a genetic as well as an environmental component. Accordingly, the results from this study suggest a molecular basis for the observed higher risk of cardiovascular disorders in PTSD patients, which raises the likelihood of cardiac dysfunction induced by long-term stress exposures.


Assuntos
Regulação da Expressão Gênica/fisiologia , Miocardite/etiologia , Miocardite/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Estresse Psicológico/complicações , Transcriptoma/fisiologia , Animais , Linhagem Celular , Proliferação de Células , Transição Epitelial-Mesenquimal/fisiologia , Matriz Extracelular/fisiologia , Perfilação da Expressão Gênica , Humanos , Estudos Longitudinais , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Análise em Microsséries , Transtornos de Estresse Pós-Traumáticos/etiologia , Estresse Psicológico/imunologia , Biologia de Sistemas
19.
Mol Biol Rep ; 43(10): 1165-78, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27510798

RESUMO

miRNAs act as important regulators of gene expression by promoting mRNA degradation or by attenuating protein translation. Since miRNAs are stably expressed in bodily fluids, there is growing interest in profiling these miRNAs, as it is minimally invasive and cost-effective as a diagnostic matrix. A technical hurdle in studying miRNA dynamics is the ability to reliably extract miRNA as small sample volumes and low RNA abundance create challenges for extraction and downstream applications. The purpose of this study was to develop a pipeline for the recovery of miRNA using small volumes of archived serum samples. The RNA was extracted employing several widely utilized RNA isolation kits/methods with and without addition of a carrier. The small RNA library preparation was carried out using Illumina TruSeq small RNA kit and sequencing was carried out using Illumina platform. A fraction of five microliters of total RNA was used for library preparation as quantification is below the detection limit. We were able to profile miRNA levels in serum from all the methods tested. We found out that addition of nucleic acid based carrier molecules had higher numbers of processed reads but it did not enhance the mapping of any miRBase annotated sequences. However, some of the extraction procedures offer certain advantages: RNA extracted by TRIzol seemed to align to the miRBase best; extractions using TRIzol with carrier yielded higher miRNA-to-small RNA ratios. Nuclease free glycogen can be carrier of choice for miRNA sequencing. Our findings illustrate that miRNA extraction and quantification is influenced by the choice of methodologies. Addition of nucleic acid- based carrier molecules during extraction procedure is not a good choice when assaying miRNA using sequencing. The careful selection of an extraction method permits the archived serum samples to become valuable resources for high-throughput applications.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , MicroRNAs/sangue , MicroRNAs/isolamento & purificação , Análise de Sequência de RNA/métodos , Coleta de Amostras Sanguíneas , Perfilação da Expressão Gênica/métodos , Biblioteca Gênica , Guanidinas , Humanos , MicroRNAs/normas , Fenóis , Kit de Reagentes para Diagnóstico
20.
BMC Genomics ; 15: 659, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25102863

RESUMO

BACKGROUND: Microgravity facilitates the opportunistic infections by augmenting the pathogenic virulence and suppressing the host resistance. Hence the extraterrestrial infections may activate potentially novel bionetworks different from the terrestrial equivalent, which could only be probed by investigating the host-pathogen relationship with a minimum of terrestrial bias. RESULTS: We customized a cell culture module to expose human endothelial cells to lipopolysaccharide (LPS). The assay was carried out onboard the STS-135 spaceflight, and a concurrent ground study constituted the baseline. Transcriptomic investigation revealed a possible immune blunting in microgravity suppressing in particular Lbp, MyD88 and MD-2, which encode proteins responsible for early LPS uptake. Certain cytokines, such as IL-6 and IL-8, surged in response to LPS insult in microgravity, as suggested by the proteomics study. Contrasting proteomic expressions of B2M, TIMP-1 and VEGRs suggested impaired pro-survival adaptation and healing mechanisms. Differential expression of miR-200a and miR-146b suggested the susceptibility of hosts in spaceflight to oxidative stress and further underscored the influence of microgravity on the immunity. CONCLUSIONS: A molecular interpretation explaining the etiology of the microgravitational impact on the host-pathogen relationship elucidated comprehensive immune blunting of the host cells responding to LPS challenges. Longer LPS exposure prompted a delayed host response, potentially ineffectual in preventing pathogens from opportunistic invasion. Significant consequences include the subsequent failure in recruiting the growth factors and a debilitated apoptosis. Follow up studies with larger sample size are warranted.


Assuntos
Células Endoteliais/metabolismo , Genômica/métodos , Lipopolissacarídeos/farmacologia , Ausência de Peso , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Análise por Conglomerados , Citocinas/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Redes Reguladoras de Genes/efeitos dos fármacos , Genoma Humano , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Oligonucleotídeos/metabolismo , Análise de Componente Principal , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Voo Espacial , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA