WHOLE BLOOD RESUSCITATION RESTORES INTESTINAL PERFUSION AND INFLUENCES GUT MICROBIOME DIVERSITY

2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Jaclyn Yracheta ◽  
Wayne Muraoka ◽  
Xiaowu Wu ◽  
David Burmeister ◽  
Daniel Darlington ◽  
...  
2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Travis T. Sims ◽  
Molly B. El Alam ◽  
Tatiana V. Karpinets ◽  
Stephanie Dorta-Estremera ◽  
Venkatesh L. Hegde ◽  
...  

AbstractDiversity of the gut microbiome is associated with higher response rates for cancer patients receiving immunotherapy but has not been investigated in patients receiving radiation therapy. Additionally, current studies investigating the gut microbiome and outcomes in cancer patients may not have adjusted for established risk factors. Here, we sought to determine if diversity and composition of the gut microbiome was independently associated with survival in cervical cancer patients receiving chemoradiation. Our study demonstrates that the diversity of gut microbiota is associated with a favorable response to chemoradiation. Additionally, compositional variation among patients correlated with short term and long-term survival. Short term survivor fecal samples were significantly enriched in Porphyromonas, Porphyromonadaceae, and Dialister, whereas long term survivor samples were significantly enriched in Escherichia Shigella, Enterobacteriaceae, and Enterobacteriales. Moreover, analysis of immune cells from cervical tumor brush samples by flow cytometry revealed that patients with a high microbiome diversity had increased tumor infiltration of CD4+ lymphocytes as well as activated subsets of CD4 cells expressing ki67+ and CD69+ over the course of radiation therapy. Modulation of the gut microbiota before chemoradiation might provide an alternative way to enhance treatment efficacy and improve treatment outcomes in cervical cancer patients.


2020 ◽  
Author(s):  
Georgina M Williams ◽  
Linda C Tapsell ◽  
Claire L O’Brien ◽  
Susan M Tosh ◽  
Eden M Barrett ◽  
...  

Abstract Context Cereal fiber modulates the gut microbiome and benefits metabolic health. The potential link between these effects is of interest.0 Objective The aim for this systematic review was to assess evidence surrounding the influence of cereal fiber intake on microbiome composition, microbiome diversity, short-chain fatty acid production, and risk factors for metabolic syndrome. Data Sources and Extraction The MEDLINE, PubMed, CINAHL, and Cochrane Library databases were searched systematically, and quality of studies was assessed using the Cochrane Risk of Bias 2.0 tool. Evidence relating to study design, dietary data collection, and outcomes was qualitatively synthesized on the basis of fiber type. Data Analysis Forty-six primary publications and 2 secondary analyses were included. Cereal fiber modulated the microbiome in most studies; however, taxonomic changes indicated high heterogeneity. Short-chain fatty acid production, microbiome diversity, and metabolic-related outcomes varied and did not always occur in parallel with microbiome changes. Poor dietary data were a further limitation. Conclusions Cereal fiber may modulate the gut microbiome; however, evidence of the link between this and metabolic outcomes is limited. Additional research is required with a focus on robust and consistent methodology. Systematic Review Registration PROSPERO registration no. CRD42018107117


Nutrients ◽  
2021 ◽  
Vol 13 (6) ◽  
pp. 1780
Author(s):  
Jean-Frédéric LeBlanc ◽  
Jonathan P. Segal ◽  
Lucia Maria de Campos Braz ◽  
Ailsa L. Hart

The gut microbiome has been implicated in a range of diseases and there is a rapidly growing understanding of this ecosystem’s importance in inflammatory bowel disease. We are yet to identify a single microbe that causes either ulcerative colitis (UC) or pouchitis, however, reduced microbiome diversity is increasingly recognised in active UC. Manipulating the gut microbiome through dietary interventions, prebiotic and probiotic compounds and faecal microbiota transplantation may expand the therapeutic landscape in UC. Specific diets, such as the Mediterranean diet or diet rich in omega-3 fatty acids, may reduce intestinal inflammation or potentially reduce the risk of incident UC. This review summarises our knowledge of gut microbiome therapies in UC and pouchitis.


Author(s):  
Anurag Singh ◽  
Davide D’Amico ◽  
Pénélope A. Andreux ◽  
Gillian Dunngalvin ◽  
Timo Kern ◽  
...  

Abstract Background Urolithin A (UA) is produced by gut microflora from foods rich in ellagitannins. UA has been shown to improve mitochondrial health preclinically and in humans. Not everyone has a microbiome capable of producing UA, making supplementation with UA an appealing strategy. Objective This is the first detailed investigation of the prevalence of UA producers in a healthy population and the ability of direct UA supplementation to overcome both microbiome and dietary variability. Dietary intake of a glass of pomegranate juice (PJ) was used to assess UA producer status (n = 100 participants) and to characterize differences in gut microbiome between UA producers from non-producers. Methods Subjects were randomized (1:1) to either PJ or a food product containing UA (500 mg). Prevalence of UA producers and non-producers were determined in the PJ group. Diet questionnaires and fecal samples were collected to compare differences between UA producers and non-producers along with plasma samples at different time points to assess levels of UA and its conjugates between the interventions. Results Only 12% of subjects had detectable levels of UA at baseline. Following PJ intake ~40% of the subjects converted significantly the precursor compounds into UA. UA producers were distinguished by a significantly higher gut microbiome diversity and ratio of Firmicutes to Bacteroides. Direct supplementation with UA significantly increased plasma levels and provided a >6-fold exposure to UA vs. PJ (p < 0.0001). Conclusions Differences in gut microbiome and diet that dictate natural exposure to UA can be overcome via direct dietary UA supplementation.


2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Tess Pallister ◽  
Matthew A. Jackson ◽  
Tiphaine C. Martin ◽  
Jonas Zierer ◽  
Amy Jennings ◽  
...  

2021 ◽  
Author(s):  
Muhammad Shahzad ◽  
Zia UlHaq ◽  
Simon C Andrews

Introduction: Tuberculosis (TB) caused by Mycobacterium tuberculosis is a common infectious disease associated with significant morbidity and mortality, especially in low and middle-income countries. Successful treatment of the disease requires prolonged intake (6 to 8 months) of multiple antibiotics with potentially detrimental consequences on the composition and functional potential of the human microbiome. The protocol described in the current study aims to identify microbiome (oral and gut) signatures associated with TB pathogenesis, treatment response and, outcome in humans. Methods and analysis: Four hundred and fifty, newly diagnosed TB patients from three district levels (Peshawar, Mardan, Swat) TB diagnosis and treatment centers will be recruited in this non-interventional, prospective cohort study and will be followed and monitored until treatment completion. Demographic and dietary intake data, anthropometric measurement and blood, stool and salivary rinse samples will be collected at baseline, day 15, month-2 and end of the treatment. Additionally, we will recruit age and sex-matched healthy controls (n=30). Blood sampling will allow monitoring of the immune response during the treatment, while salivary rinse and fecal samples will allow monitoring of dynamic changes in oral and gut microbiome diversity. Within this prospective cohort study, a nested case-control study design will be conducted to assess perturbations in oral and gut microbiome diversity (microbial dysbiosis) and immune response and compare between the patients groups (treatment success vs failure). Ethics and dissemination The study has received ethics approval from the Ethic Board of Khyber Medical University Peshawar, and administrative approval from the Provincial TB Control Program of Khyber Pakhtunkhwa, Pakistan. The study results will be presented at national and international conferences and published in peer-reviewed journals. Trial registration number: NCT04985994; pre-results


2021 ◽  
Author(s):  
Aoji Xie ◽  
Elizabeth Ensink ◽  
Peipei Li ◽  
Juozas Gordevicius ◽  
Lee L. Marshall ◽  
...  

Background The gut microbiome and its metabolites can impact brain health and are altered in Parkinson's disease (PD) patients. It has been recently demonstrated that PD patients have reduced fecal levels of the potent epigenetic modulator butyrate and its bacterial producers. Here, we investigate whether the changes in the gut microbiome and associated metabolites are linked to PD symptoms and epigenetic markers in leucocytes and neurons. Methods Stool, whole blood samples, and clinical data were collected from 55 PD patients and 55 controls. We performed DNA methylation analysis on whole blood samples and analyzed the results in relation to fecal short-chain fatty acid concentrations and microbiota composition. In another cohort, prefrontal cortex neurons were isolated from control and PD brains. We identified the genome-wide DNA methylation by targeted bisulfite sequencing. Results We show that lower fecal butyrate and reduced Roseburia, Romboutsia, and Prevotella counts are linked to depressive symptoms in PD patients. Genes containing butyrate-associated methylation sites include PD risk genes and significantly overlap with sites epigenetically altered in PD blood leucocytes, predominantly neutrophils, and in brain neurons, relative to controls. Moreover, butyrate-associated methylated-DNA (mDNA) regions in PD overlap with those altered in gastrointestinal, autoimmune, and psychiatric diseases.


2013 ◽  
Vol 27 (S1) ◽  
Author(s):  
Nanette Steinle ◽  
Shana Cirimotch ◽  
Kathy Ryan ◽  
Claire Fraser ◽  
Alan Shuldiner ◽  
...  

2021 ◽  
Author(s):  
Lauren Petrullo ◽  
Tiantian Ren ◽  
Martin Wu ◽  
Rudy Boonstra ◽  
Rupert Palme ◽  
...  

Gut microbiome diversity plays an important role in host health and fitness, in part through the diversification of gut metabolic function and pathogen protection. Elevations in glucocorticoids (GCs) appear to reduce gut microbiome diversity in experimental studies, suggesting that a loss of microbial diversity may be a negative consequence of increased GCs. However, given that ecological factors like food availability and population density may independently influence both GCs and microbial diversity, understanding how these factors structure the GC-microbiome relationship is crucial to interpreting its significance in wild populations. Here, we used an ecological framework to investigate the relationship between GCs and gut microbiome diversity in wild North American red squirrels (Tamiasciurus hudsonicus). We found that higher GCs predicted lower gut microbiome diversity and an increase in metabolic taxa. In addition, we identified a loss of potentially pathogenic bacteria with increasing GCs. Both dietary heterogeneity and an upcoming masting event exhibited direct effects on gut microbiome diversity, whereas conspecific density and host reproductive activity impacted diversity indirectly via changes in GCs. Together, our results suggest that GCs coordinate the effects of ecological change and host biology on gut microbiome diversity, and highlight the importance of situating the GC-microbiome relationship within an ecological framework.


Sign in / Sign up

Export Citation Format

Share Document