Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 272
Filter
1.
Cell ; 186(22): 4851-4867.e20, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37848036

ABSTRACT

Post-acute sequelae of COVID-19 (PASC, "Long COVID") pose a significant global health challenge. The pathophysiology is unknown, and no effective treatments have been found to date. Several hypotheses have been formulated to explain the etiology of PASC, including viral persistence, chronic inflammation, hypercoagulability, and autonomic dysfunction. Here, we propose a mechanism that links all four hypotheses in a single pathway and provides actionable insights for therapeutic interventions. We find that PASC are associated with serotonin reduction. Viral infection and type I interferon-driven inflammation reduce serotonin through three mechanisms: diminished intestinal absorption of the serotonin precursor tryptophan; platelet hyperactivation and thrombocytopenia, which impacts serotonin storage; and enhanced MAO-mediated serotonin turnover. Peripheral serotonin reduction, in turn, impedes the activity of the vagus nerve and thereby impairs hippocampal responses and memory. These findings provide a possible explanation for neurocognitive symptoms associated with viral persistence in Long COVID, which may extend to other post-viral syndromes.


Subject(s)
Post-Acute COVID-19 Syndrome , Serotonin , Humans , COVID-19/complications , Disease Progression , Inflammation , Post-Acute COVID-19 Syndrome/blood , Post-Acute COVID-19 Syndrome/pathology , Serotonin/blood , Virus Diseases
2.
Cell ; 167(6): 1469-1480.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912057

ABSTRACT

The intestinal microbiota influence neurodevelopment, modulate behavior, and contribute to neurological disorders. However, a functional link between gut bacteria and neurodegenerative diseases remains unexplored. Synucleinopathies are characterized by aggregation of the protein α-synuclein (αSyn), often resulting in motor dysfunction as exemplified by Parkinson's disease (PD). Using mice that overexpress αSyn, we report herein that gut microbiota are required for motor deficits, microglia activation, and αSyn pathology. Antibiotic treatment ameliorates, while microbial re-colonization promotes, pathophysiology in adult animals, suggesting that postnatal signaling between the gut and the brain modulates disease. Indeed, oral administration of specific microbial metabolites to germ-free mice promotes neuroinflammation and motor symptoms. Remarkably, colonization of αSyn-overexpressing mice with microbiota from PD-affected patients enhances physical impairments compared to microbiota transplants from healthy human donors. These findings reveal that gut bacteria regulate movement disorders in mice and suggest that alterations in the human microbiome represent a risk factor for PD.


Subject(s)
Parkinson Disease/microbiology , Parkinson Disease/pathology , Animals , Brain/pathology , Dysbiosis/pathology , Fatty Acids/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice , Microglia/pathology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , alpha-Synuclein/metabolism
3.
Cell ; 160(3): 447-60, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619688

ABSTRACT

Decreases in the diversity of enteric bacterial populations are observed in patients with Crohn's disease (CD) and ulcerative colitis (UC). Less is known about the virome in these diseases. We show that the enteric virome is abnormal in CD and UC patients. In-depth analysis of preparations enriched for free virions in the intestine revealed that CD and UC were associated with a significant expansion of Caudovirales bacteriophages. The viromes of CD and UC patients were disease and cohort specific. Importantly, it did not appear that expansion and diversification of the enteric virome was secondary to changes in bacterial populations. These data support a model in which changes in the virome may contribute to intestinal inflammation and bacterial dysbiosis. We conclude that the virome is a candidate for contributing to, or being a biomarker for, human inflammatory bowel disease and speculate that the enteric virome may play a role in other diseases.


Subject(s)
Caudovirales/isolation & purification , Colitis, Ulcerative/virology , Crohn Disease/virology , Dysbiosis/virology , Microviridae/isolation & purification , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Caudovirales/genetics , Cohort Studies , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/therapy , Crohn Disease/microbiology , Crohn Disease/pathology , Crohn Disease/therapy , Dysbiosis/microbiology , Dysbiosis/pathology , Dysbiosis/therapy , Feces/microbiology , Feces/virology , Humans , Metagenome , Microviridae/genetics
4.
J Infect Dis ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39011957

ABSTRACT

Host metabolic dysregulation, especially in tryptophan metabolism, is intricately linked to COVID-19 severity and its post-acute sequelae (Long COVID). People living with HIV (PLWH) experience similar metabolic dysregulation and face an increased risk of developing Long COVID. However, whether pre-existing HIV-associated metabolic dysregulations contribute in predisposing PLWH to severe COVID-19 outcomes remains underexplored. Analyzing pre-pandemic samples from PLWH with documented post-infection outcomes, we found specific metabolic alterations, including increased tryptophan catabolism, predicting an elevated risk of severe COVID-19 and the incidence of Long COVID. These alterations warrant further investigation for their potential prognostic and mechanistic significance in determining COVID-19 complications.

5.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161260

ABSTRACT

Individuals who are minoritized as a result of race, sexual identity, gender, or socioeconomic status experience a higher prevalence of many diseases. Understanding the biological processes that cause and maintain these socially driven health inequities is essential for addressing them. The gut microbiome is strongly shaped by host environments and affects host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities. Nevertheless, few studies have directly integrated the gut microbiome into investigations of health inequities. Here, we argue that accounting for host-gut microbe interactions will improve understanding and management of health inequities, and that health policy must begin to consider the microbiome as an important pathway linking environments to population health.


Subject(s)
Gastrointestinal Microbiome , Health Status Disparities , Disease , Health , Humans , Mental Health , Publications
6.
Proteomics ; 23(19): e2300023, 2023 10.
Article in English | MEDLINE | ID: mdl-37525324

ABSTRACT

The epidemy of metabolic syndrome (MetS) is typically preceded by adoption of a "risky" lifestyle (e.g., dietary habit) among populations. Evidence shows that those with low socioeconomic status (SES) are at an increased risk for MetS. To investigate this, we recruited 123 obese subjects (body mass index [BMI] ≥ 30) from Chicago. Multi-omic data were collected to interrogate fecal microbiota, systemic markers of inflammation and immune activation, plasma metabolites, and plasma glycans. Intestinal permeability was measured using the sugar permeability testing. Our results suggest a heterogenous metabolic dysregulation among obese populations who are at risk of MetS. Systemic inflammation, linked to poor diet, intestinal microbiome dysbiosis, and gut barrier dysfunction may explain the development of MetS in these individuals. Our analysis revealed 37 key features associated with increased numbers of MetS features. These features were used to construct a composite metabolic-inflammatory (MI) score that was able to predict progression of MetS among at-risk individuals. The MI score was correlated with several markers of poor diet quality as well as lower levels of gut microbial diversity and abnormalities in several species of bacteria. This study reveals novel targets to reduce the burden of MetS and suggests access to healthy food options as a practical intervention.


Subject(s)
Metabolic Syndrome , Microbiota , Humans , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Multiomics , Socioeconomic Disparities in Health , Diet , Obesity/metabolism , Inflammation , Dysbiosis/complications , Dysbiosis/microbiology
7.
Mov Disord ; 38(3): 399-409, 2023 03.
Article in English | MEDLINE | ID: mdl-36691982

ABSTRACT

BACKGROUND: The gut microbiome is altered in several neurologic disorders, including Parkinson's disease (PD). OBJECTIVES: The aim is to profile the fecal gut metagenome in PD for alterations in microbial composition, taxon abundance, metabolic pathways, and microbial gene products, and their relationship with disease progression. METHODS: Shotgun metagenomic sequencing was conducted on 244 stool donors from two independent cohorts in the United States, including individuals with PD (n = 48, n = 47, respectively), environmental household controls (HC, n = 29, n = 30), and community population controls (PC, n = 41, n = 49). Microbial features consistently altered in PD compared to HC and PC subjects were identified. Data were cross-referenced to public metagenomic data sets from two previous studies in Germany and China to determine generalizable microbiome features. RESULTS: We find several significantly altered taxa between PD and controls within the two cohorts sequenced in this study. Analysis across global cohorts returns consistent changes only in Intestinimonas butyriciproducens. Pathway enrichment analysis reveals disruptions in microbial carbohydrate and lipid metabolism and increased amino acid and nucleotide metabolism in PD. Global gene-level signatures indicate an increased response to oxidative stress, decreased cellular growth and microbial motility, and disrupted intercommunity signaling. CONCLUSIONS: A metagenomic meta-analysis of PD shows consistent and novel alterations in functional metabolic potential and microbial gene abundance across four independent studies from three continents. These data reveal that stereotypic changes in the functional potential of the gut microbiome are a consistent feature of PD, highlighting potential diagnostic and therapeutic avenues for future research. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Gastrointestinal Microbiome , Parkinson Disease , Humans , Parkinson Disease/diagnosis , Metagenome/genetics , Cohort Studies , Gastrointestinal Microbiome/genetics , Feces
8.
FASEB J ; 36(1): e22043, 2022 01.
Article in English | MEDLINE | ID: mdl-34861073

ABSTRACT

Circadian misalignment-the misalignment between the central circadian "clock" and behavioral and environmental cycles (including sleep/wake, fasting/eating, dark/light)-results in adverse cardiovascular and metabolic effects. Potential underlying mechanisms for these adverse effects include alterations in the orogastrointestinal microbiota. However, it remains unknown whether human oral microbiota has endogenous circadian rhythms (i.e., independent of sleep/wake, fasting/eating, and dark/light cycles) and whether circadian misalignment influences oral microbiota community composition. Healthy young individuals [27.3 ± 2.3 years (18-35 years), 4 men and 2 women, body-mass index range: 18-28 kg/m2 ] were enrolled in a stringently controlled 14-day circadian laboratory protocol. This included a 32-h constant routine (CR) protocol (endogenous circadian baseline assessment), a forced desynchrony protocol with four 28-h "days" under ~3 lx to induce circadian misalignment, and a post-misalignment 40-h CR protocol. Microbiota assessments were performed on saliva samples collected every 4 h throughout both CR protocols. Total DNA was extracted and processed using high-throughput 16S ribosomal RNA gene amplicon sequencing. The relative abundance of specific oral microbiota populations, i.e., one of the five dominant phyla, and three of the fourteen dominant genera, exhibited significant endogenous circadian rhythms. Importantly, circadian misalignment dramatically altered the oral microbiota landscape, such that four of the five dominant phyla and eight of the fourteen dominant genera exhibited significant circadian misalignment effects. Moreover, circadian misalignment significantly affected the metagenome functional content of oral microbiota (inferred gene content analysis), as indicated by changes in specific functional pathways associated with metabolic control and immunity. Collectively, our proof-of-concept study provides evidence for endogenous circadian rhythms in human oral microbiota and show that even relatively short-term experimental circadian misalignment can dramatically affect microbiota community composition and functional pathways involved in metabolism and immune function. These proof-of-principle findings have translational relevance to individuals typically exposed to circadian misalignment, including night shift workers and frequent flyers.


Subject(s)
Circadian Rhythm , Microbiota , Mouth/microbiology , Saliva/microbiology , Shift Work Schedule , Adolescent , Adult , Female , Humans , Male , Proof of Concept Study
9.
Eur J Neurol ; 30(11): 3581-3594, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36593694

ABSTRACT

BACKGROUND AND PURPOSE: The role of the gut microbiome in the pathogenesis of Parkinson disease (PD) is under intense investigation, and the results presented are still very heterogeneous. These discrepancies arise not only from the highly heterogeneous pathology of PD, but also from widely varying methodologies at all stages of the workflow, from sampling to final statistical analysis. The aim of the present work is to harmonize the workflow across studies to reduce the methodological heterogeneity and to perform a pooled analysis to account for other sources of heterogeneity. METHODS: We performed a systematic review to identify studies comparing the gut microbiota of PD patients to healthy controls. A workflow was designed to harmonize processing across all studies from bioinformatics processing to final statistical analysis using a Bayesian random-effects meta-analysis based on individual patient-level data. RESULTS: The results show that harmonizing workflows minimizes differences between statistical methods and reveals only a small set of taxa being associated with the pathogenesis of PD. Increased shares of the genera Akkermansia and Bifidobacterium and decreased shares of the genera Roseburia and Faecalibacterium were most characteristic for PD-associated microbiota. CONCLUSIONS: Our study summarizes evidence that reduced levels of butyrate-producing taxa in combination with possible degradation of the mucus layer by Akkermansia may promote intestinal inflammation and reduced permeability of the gut mucosal layer. This may allow potentially pathogenic metabolites to transit and enter the enteric nervous system.

10.
Semin Neurol ; 43(4): 506-517, 2023 08.
Article in English | MEDLINE | ID: mdl-37562457

ABSTRACT

The gut-brain axis refers to a bidirectional communication pathway linking the gastrointestinal system to the central nervous system. The hardware of this multifaceted pathway takes many forms, at once structural (neurons, microglia, intestinal epithelial cell barrier), chemical (neurotransmitters, enteroendocrine hormones, bacterial metabolites), and cellular (immune signaling, inflammatory pathways). The gut-brain axis is exquisitely influenced by our environment, diet, and behaviors. Here, we will describe recent progress in understanding the gut-brain axis in neurological disease, using Parkinson's disease as a guide. We will see that each component of the gut-brain axis is heavily mediated by intestinal microbiota and learn how gut-brain communication can go awry in microbial dysbiosis.


Subject(s)
Gastrointestinal Microbiome , Parkinson Disease , Humans , Brain-Gut Axis , Brain , Central Nervous System , Parkinson Disease/metabolism , Gastrointestinal Microbiome/physiology
11.
Am J Physiol Lung Cell Mol Physiol ; 323(3): L266-L280, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35699290

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is often associated with intestinal comorbidities. In this study, changes in intestinal homeostasis and immunity in a cigarette smoke (CS)- and lipopolysaccharide (LPS)-induced COPD model were investigated. Mice were exposed to cigarette smoke or air for 72 days, except days 42, 52, and 62 on which the mice were treated with saline or LPS via intratracheal instillation. Cigarette smoke exposure increased the airway inflammatory cell numbers, mucus production, and different inflammatory mediators, including C-reactive protein (CRP) and keratinocyte-derived chemokine (KC), in bronchoalveolar lavage (BAL) fluid and serum. LPS did not further impact airway inflammatory cell numbers or mucus production but decreased inflammatory mediator levels in BAL fluid. T helper (Th) 1 cells were enhanced in the spleen after cigarette smoke exposure; however, in combination with LPS, cigarette exposure caused an increase in Th1 and Th2 cells. Histomorphological changes were observed in the proximal small intestine after cigarette smoke exposure, and addition of LPS had no effect. Cigarette smoke activated the intestinal immune network for IgA production in the distal small intestine that was associated with increased fecal sIgA levels and enlargement of Peyer's patches. Cigarette smoke plus LPS decreased fecal sIgA levels and the size of Peyer's patches. In conclusion, cigarette smoke with or without LPS affects intestinal health as observed by changes in intestinal histomorphology and immune network for IgA production. Elevated systemic mediators might play a role in the lung-gut cross talk. These findings contribute to a better understanding of intestinal disorders related to COPD.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Animals , Bronchoalveolar Lavage Fluid , Cigarette Smoking/adverse effects , Disease Models, Animal , Homeostasis , Immunoglobulin A/adverse effects , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/metabolism , Immunoglobulin A, Secretory/pharmacology , Lipopolysaccharides/adverse effects , Lung/metabolism , Mice , Pulmonary Disease, Chronic Obstructive/metabolism , Nicotiana
12.
Neurobiol Dis ; 170: 105780, 2022 08.
Article in English | MEDLINE | ID: mdl-35654277

ABSTRACT

There is growing appreciation of the importance of the intestinal microbiota in Parkinson's disease (PD), and one potential mechanism by which the intestinal microbiota can communicate with the brain is via bacteria-derived metabolites. In this study, plasma levels of bacterial-derived metabolites including trimethylamine-N-oxide (TMAO), short chain fatty acids (SCFA), the branched chain fatty acid isovalerate, succinate, and lactate were evaluated in PD subjects (treatment naïve and treated) which were compared to (1) population controls, (2) spousal / household controls (similar lifestyle to PD subjects), and (3) subjects with multiple system atrophy (MSA). Analyses revealed an increase in the TMAO pathway in PD subjects which was independent of medication status, disease characteristics, and lifestyle. Lactic acid was decreased in treated PD subjects, succinic acid positively correlated with disease severity, and the ratio of pro-inflammatory TMAO to the putative anti-inflammatory metabolite butyric acid was significantly higher in PD subjects compared to controls indicating a pro-inflammatory shift in the metabolite profile in PD subjects. Finally, acetic and butyric acid were different between PD and MSA subjects indicating that metabolites may differentiate these synucleinopathies. In summary, (1) TMAO is elevated in PD subjects, a phenomenon independent of disease characteristics, treatment status, and lifestyle and (2) metabolites may differentiate PD and MSA subjects. Additional studies to understand the potential of TMAO and other bacterial metabolites to serve as a biomarker or therapeutic targets are warranted.


Subject(s)
Gastrointestinal Microbiome , Multiple System Atrophy , Parkinson Disease , Bacteria , Butyrates , Humans , Life Style , Parkinson Disease/therapy
13.
Am J Physiol Gastrointest Liver Physiol ; 323(2): G61-G70, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35638693

ABSTRACT

Posttraumatic stress disorder (PTSD) is a psychiatric disorder, resulting from exposure to traumatic events. Current recommended first-line interventions for the treatment of PTSD include evidence-based psychotherapies, such as cognitive processing therapy (CPT). Psychotherapies are effective for reducing PTSD symptoms, but approximately two-thirds of veterans continue to meet diagnostic criteria for PTSD after treatment, suggesting there is an incomplete understanding of what factors sustain PTSD. The intestine can influence the brain and this study evaluated intestinal readouts in subjects with PTSD. Serum samples from controls without PTSD (n = 40) from the Duke INTRuST Program were compared with serum samples from veterans with PTSD (n = 40) recruited from the Road Home Program at Rush University Medical Center. Assessments included microbial metabolites, intestinal barrier, and intestinal epithelial cell function. In addition, intestinal readouts were assessed in subjects with PTSD before and after a 3-wk CPT-based intensive treatment program (ITP) to understand if treatment impacts the intestine. Compared with controls, veterans with PTSD had a proinflammatory intestinal environment including lower levels of microbiota-derived metabolites, such as acetic, lactic, and succinic acid, intestinal barrier dysfunction [lipopolysaccharide (LPS) and LPS-binding protein], an increase in HMGB1, and a concurrent increase in the number of intestinal epithelial cell-derived extracellular vesicles. The ITP improved PTSD symptoms but no changes in intestinal outcomes were noted. This study confirms the intestine is abnormal in subjects with PTSD and suggests that effective treatment of PTSD does not alter intestinal readouts. Targeting beneficial changes in the intestine may be an approach to enhance existing PTSD treatments.NEW & NOTEWORTHY This study confirms an abnormal intestinal environment is present in subjects with PTSD. This study adds to what is already known by examining the intestinal barrier and evaluating the relationship between intestinal readouts and PTSD symptoms and is the first to report the impact of PTSD treatment (which improves symptoms) on intestinal readouts. This study suggests that targeting the intestine as an adjunct approach could improve the treatment of PTSD.


Subject(s)
Cognitive Behavioral Therapy , Stress Disorders, Post-Traumatic , Veterans , Cognitive Behavioral Therapy/methods , Humans , Intestines , Stress Disorders, Post-Traumatic/therapy , Treatment Outcome , Veterans/psychology
14.
Alcohol Clin Exp Res ; 46(11): 1930-1943, 2022 11.
Article in English | MEDLINE | ID: mdl-36394508

ABSTRACT

In Dec. 2019-January 2020, a pneumonia illness originating in Wuhan, China, designated as coronavirus disease 2019 (COVID-19) was shown to be caused by a novel RNA coronavirus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). People with advanced age, male sex, and/or underlying health conditions (obesity, type 2 diabetes, cardiovascular disease, hypertension, chronic kidney disease, and chronic lung disease) are especially vulnerable to severe COVID-19 symptoms and death. These risk factors impact the immune system and are also associated with poor health, chronic illness, and shortened longevity. However, a large percent of patients without these known risk factors also develops severe COVID-19 disease that can result in death. Thus, there must exist risk factors that promote exaggerated inflammatory and immune response to the SARS-CoV-2 virus leading to death. One such risk factor may be alcohol misuse and alcohol use disorder because these can exacerbate viral lung infections like SARS, influenza, and pneumonia. Thus, it is highly plausible that alcohol misuse is a risk factor for either increased infection rate when individuals are exposed to SARS-CoV-2 virus and/or more severe COVID-19 in infected patients. Alcohol use is a well-known risk factor for lung diseases and ARDS in SARS patients. We propose that alcohol has three key pathogenic elements in common with other COVID-19 severity risk factors: namely, inflammatory microbiota dysbiosis, leaky gut, and systemic activation of the NLRP3 inflammasome. We also propose that these three elements represent targets for therapy for severe COVID-19.


Subject(s)
Alcoholism , COVID-19 , Diabetes Mellitus, Type 2 , Humans , Male , Alcoholism/epidemiology , SARS-CoV-2 , Risk Factors , Ethanol
15.
Curr Neurol Neurosci Rep ; 22(7): 335-342, 2022 07.
Article in English | MEDLINE | ID: mdl-35633466

ABSTRACT

PURPOSE OF REVIEW: Defective gut-brain communication has recently been proposed as a promoter of neurodegeneration, but mechanisms mediating communication remain elusive. In particular, the Parkinson's disease (PD) phenotype has been associated with both dysbiosis of intestinal microbiota and neuroinflammation. Here, we review recent advances in the PD field that connect these two concepts, providing an explanation based on enteroendocrine signaling from the gut to the brain. RECENT FINDINGS: There have been several recent accounts highlighting the importance of the microbiota-gut-brain axis in PD. The objective of this review is to discuss the role of the neuroendocrine system in gut-brain communication as it relates to PD pathogenesis, as this system has not been comprehensively considered in prior reviews. The incretin hormone glucagon-like peptide 1 (GLP-1) is secreted by enteroendocrine cells of the intestinal epithelium, and there is evidence that it is neuroprotective in animal models and human subjects with PD. Agonists of GLP-1 receptors used in diabetes appear to be useful for preventing neurodegeneration. New tools and models have enabled us to study regulation of GLP-1 secretion by intestinal microbiota, to understand how this process may be defective in PD, and to develop methods for therapeutically modifying disease development or progression using the enteroendocrine system. GLP-1 secretion by enteroendocrine cells may be a key mediator of neuroprotection in PD, and new findings in this field may offer unique insights into PD pathogenesis and therapeutic strategies.


Subject(s)
Brain-Gut Axis , Glucagon-Like Peptide 1 , Neurosecretory Systems , Parkinson Disease , Animals , Brain/pathology , Dysbiosis , Glucagon-Like Peptide 1/physiology , Humans , Neurosecretory Systems/physiology , Parkinson Disease/physiopathology
16.
Eat Weight Disord ; 27(4): 1395-1403, 2022 May.
Article in English | MEDLINE | ID: mdl-34355307

ABSTRACT

BACKGROUND: Metabolic dysfunction and obesity rates are on the rise. Although the central modes of circadian disruption has been studied in relation to the risk of obesity, the role of eating time has remained unclear. Here, we aimed to assess circadian behavioral phenotypes and their association with the risk of elevated body mass index (BMI). METHODS: This was a prospective cross-sectional study of individuals presenting for colorectal cancer screening colonoscopy. Participants completed demographic questionnaires, The Munich ChronoType Questionnaire (MCTQ), and Food Timing Screener (FTS). The primary outcome of the study was the association between circadian phenotypes and elevated BMI. RESULTS: A total of 488 individuals completed the survey, with a mean (SD) age of 57.5 (10.8) years. The mean body mass index (BMI) was 28.8 (6.1) kg/m2, with 72.3% of individuals met criteria for elevated BMI. Four circadian behavioral phenotypes were generated: early chronotype with regular food timing (ER) (34.7%), early chronotype with irregular food timing (EI) (11.7%), intermediate/late chronotype with regular food timing (LR) (33.9%), and intermediate/late chronotype with irregular food timing (LI) (19.7%). In a multivariable regression analysis, LI phenotype had 2.9 times higher odds of elevated BMI as compared to ER phenotype (OR 2.9, 95% CI 1.3-6.7, P = 0.01). CONCLUSION: The combination of late chronotype and irregular food timing, representative of a behavioral circadian rhythm disruption, is associated with higher rates of elevated BMI. The majority of individuals with this abnormal circadian phenotype were younger than 60 years old. This observation is especially relevant because of the ongoing rise in the obesity rates among young adults. LEVEL III: Evidence obtained from well-designed cohort or case-control analytic studies.


Subject(s)
Circadian Rhythm , Sleep , Body Mass Index , Cross-Sectional Studies , Humans , Obesity , Phenotype , Prospective Studies , Surveys and Questionnaires
17.
Biol Proced Online ; 23(1): 10, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34058978

ABSTRACT

We investigated nasopharyngeal microbial community structure in COVID-19-positive and -negative patients. High-throughput 16S ribosomal RNA gene amplicon sequencing revealed significant microbial community structure differences between COVID-19-positive and -negative patients. This proof-of-concept study demonstrates that: (1) nasopharyngeal microbiome communities can be assessed using collection samples already collected for SARS-CoV-2 testing (viral transport media) and (2) SARS-CoV-2 infection is associated with altered dysbiotic microbial profiles which could be a biomarker for disease progression and prognosis in SARS-CoV-2.

18.
FASEB J ; 34(11): 14302-14317, 2020 11.
Article in English | MEDLINE | ID: mdl-32931052

ABSTRACT

Particles released from implants cause inflammatory bone loss, which is a key factor in aseptic loosening, the most common reason for joint replacement failure. With the anticipated increased incidence of total joint replacement in the next decade, implant failure will continue to burden patients. The gut microbiome is increasingly recognized as an important factor in bone physiology, however, its role in implant loosening is currently unknown. We tested the hypothesis that implant loosening is associated with changes in the gut microbiota in a preclinical model. When the particle challenge caused local joint inflammation, decreased peri-implant bone volume, and decreased implant fixation, the gut microbiota was affected. When the particle challenge did not cause this triad of local effects, the gut microbiota was not affected. Our results suggest that cross-talk between these compartments is a previously unrecognized mechanism of failure following total joint replacement.


Subject(s)
Gastrointestinal Microbiome , Inflammation/pathology , Osteolysis/pathology , Prostheses and Implants/adverse effects , Prosthesis-Related Infections/pathology , Animals , Inflammation/etiology , Male , Osteolysis/etiology , Prosthesis-Related Infections/etiology , Rats
19.
Pediatr Allergy Immunol ; 32(5): 1006-1012, 2021 07.
Article in English | MEDLINE | ID: mdl-33570236

ABSTRACT

BACKGROUND: Previous studies have shown that a child's risk of developing atopic disease is impacted by both genetic and environmental factors. Because small children spend the majority of their time in their homes, exposure to microbial factors in their home environment may be protective or risk factors for development of atopic diseases, such as atopic dermatitis. METHODS: Dust samples from the homes of 86 Black South African children 12 to 36 months old were collected for analysis of the bacterial microbiome. This case-control study design included children with and without atopic dermatitis from rural and urban environments. RESULTS: Significant differences in bacterial composition and diversity were found when comparing children with and without atopic dermatitis. Furthermore, house dust microbiota was significantly different in rural and urban areas. Differences were best accounted for by higher relative abundance of Ruminococcaceae, Lachnospiraceae, and Bacteroidaceae families in rural compared with urban houses. Levels of Ruminococcaceae were also found to be significantly depleted in the house dust of rural children with atopic dermatitis as compared to control children. CONCLUSIONS: House dust composition may be an important risk factor for the development of atopic disease, and this association may be driven in part by the gut microbiome. Low levels of the Ruminococcaceae family from Clostridia class in particular may explain the association between urban living and atopy. However, further research is needed to elucidate these links.


Subject(s)
Dermatitis, Atopic , Microbiota , Case-Control Studies , Child , Child, Preschool , Dermatitis, Atopic/epidemiology , Dust , Humans , Infant , Urbanization
20.
BMC Biol ; 18(1): 137, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33076930

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Gastrointestinal tract dysfunction is one of the non-motor features, where constipation is reported as the most common gastrointestinal symptom. Aromatic bacterial metabolites are attracting considerable attention due to their impact on gut homeostasis and host's physiology. In particular, Clostridium sporogenes is a key contributor to the production of these bioactive metabolites in the human gut. RESULTS: Here, we show that C. sporogenes deaminates levodopa, the main treatment in Parkinson's disease, and identify the aromatic aminotransferase responsible for the initiation of the deamination pathway. The deaminated metabolite from levodopa, 3-(3,4-dihydroxyphenyl)propionic acid, elicits an inhibitory effect on ileal motility in an ex vivo model. We detected 3-(3,4-dihydroxyphenyl)propionic acid in fecal samples of Parkinson's disease patients on levodopa medication and found that this metabolite is actively produced by the gut microbiota in those stool samples. CONCLUSIONS: Levodopa is deaminated by the gut bacterium C. sporogenes producing a metabolite that inhibits ileal motility ex vivo. Overall, this study underpins the importance of the metabolic pathways of the gut microbiome involved in drug metabolism not only to preserve drug effectiveness, but also to avoid potential side effects of bacterial breakdown products of the unabsorbed residue of medication.


Subject(s)
Antiparkinson Agents/metabolism , Clostridium/metabolism , Gastrointestinal Motility , Levodopa/metabolism , Transaminases/metabolism , Animals , Antiparkinson Agents/chemistry , Clostridium/enzymology , Deamination , Gastrointestinal Microbiome , Levodopa/chemistry , Male , Mice/microbiology , Mice, Inbred C57BL , Parkinson Disease/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL