Nature Microbiology, volume 7, issue 5, pages 630-639

Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in women

Colin J. Worby 1
Henry L. Schreiber 2, 3, 4
TIMOTHY STRAUB 1
Lucas R Van Dijk 1, 5
Ryan A Bronson 1
Benjamin S. Olson 2, 3
Jerome S. Pinkner 2
Chloe L P Obernuefemann 2
Vanessa L Muñoz 2
Alexandra E Paharik 2
Philippe N Azimzadeh 2
Bruce J Walker 6
Christopher A Desjardins 1
Wen-Chi Chou 1
Karla Bergeron 7
Sinéad B Chapman 1
Aleksandra Klim 7
Abigail L. Manson 1
Thomas J. Hannan 8
Thomas M. Hooton 9
Andrew L. Kau 3, 10
H. Henry Lai 7, 11
Karen W. Dodson 2, 3
S.J. Hultgren 2, 3
Show full list: 25 authors
Publication typeJournal Article
Publication date2022-05-03
scimago Q1
SJR7.982
CiteScore44.4
Impact factor20.5
ISSN20585276
Cell Biology
Genetics
Microbiology (medical)
Microbiology
Applied Microbiology and Biotechnology
Immunology
Abstract
Recurrent urinary tract infections (rUTIs) are a major health burden worldwide, with history of infection being a significant risk factor. While the gut is a known reservoir for uropathogenic bacteria, the role of the microbiota in rUTI remains unclear. We conducted a year-long study of women with (n = 15) and without (n = 16) history of rUTI, from whom we collected urine, blood and monthly faecal samples for metagenomic and transcriptomic interrogation. During the study 24 UTIs were reported, with additional samples collected during and after infection. The gut microbiome of individuals with a history of rUTI was significantly depleted in microbial richness and butyrate-producing bacteria compared with controls, reminiscent of other inflammatory conditions. However, Escherichia coli gut and bladder populations were comparable between cohorts in both relative abundance and phylogroup. Transcriptional analysis of peripheral blood mononuclear cells revealed expression profiles indicative of differential systemic immunity between cohorts. Altogether, these results suggest that rUTI susceptibility is in part mediated through the gut–bladder axis, comprising gut dysbiosis and differential immune response to bacterial bladder colonization, manifesting in symptoms. Multi-omics analyses of faecal, urine and blood samples from women with and without recurrent urinary tract infections reveal that gut dysbiosis and differential immune responses may play a role in risk of infection via the gut–bladder axis.
van Dijk L.R., Walker B.J., Straub T.J., Worby C.J., Grote A., Schreiber H.L., Anyansi C., Pickering A.J., Hultgren S.J., Manson A.L., Abeel T., Earl A.M.
Genome Biology scimago Q1 wos Q1 Open Access
2022-03-07 citations by CoLab: 65 Abstract  
Human-associated microbial communities comprise not only complex mixtures of bacterial species, but also mixtures of conspecific strains, the implications of which are mostly unknown since strain level dynamics are underexplored due to the difficulties of studying them. We introduce the Strain Genome Explorer (StrainGE) toolkit, which deconvolves strain mixtures and characterizes component strains at the nucleotide level from short-read metagenomic sequencing with higher sensitivity and resolution than other tools. StrainGE is able to identify strains at 0.1x coverage and detect variants for multiple conspecific strains within a sample from coverages as low as 0.5x.
Asnicar F., Leeming E.R., Dimidi E., Mazidi M., Franks P.W., Al Khatib H., Valdes A.M., Davies R., Bakker E., Francis L., Chan A., Gibson R., Hadjigeorgiou G., Wolf J., Spector T.D., et. al.
Gut scimago Q1 wos Q1
2021-03-15 citations by CoLab: 122 Abstract  
Background and aimsGut transit time is a key modulator of host–microbiome interactions, yet this is often overlooked, partly because reliable methods are typically expensive or burdensome. The aim of this single-arm, single-blinded intervention study is to assess (1) the relationship between gut transit time and the human gut microbiome, and (2) the utility of the ‘blue dye’ method as an inexpensive and scalable technique to measure transit time.MethodsWe assessed interactions between the taxonomic and functional potential profiles of the gut microbiome (profiled via shotgun metagenomic sequencing), gut transit time (measured via the blue dye method), cardiometabolic health and diet in 863 healthy individuals from the PREDICT 1 study.ResultsWe found that gut microbiome taxonomic composition can accurately discriminate between gut transit time classes (0.82 area under the receiver operating characteristic curve) and longer gut transit time is linked with specific microbial species such as Akkermansia muciniphila, Bacteroides spp and Alistipes spp (false discovery rate-adjusted p values <0.01). The blue dye measure of gut transit time had the strongest association with the gut microbiome over typical transit time proxies such as stool consistency and frequency.ConclusionsGut transit time, measured via the blue dye method, is a more informative marker of gut microbiome function than traditional measures of stool consistency and frequency. The blue dye method can be applied in large-scale epidemiological studies to advance diet-microbiome-health research. Clinical trial registry website https://clinicaltrials.gov/ct2/show/NCT03479866 and trial number NCT03479866.
de Santiago P.R., Blanco A., Morales F., Marcelain K., Harismendy O., Sjöberg Herrera M., Armisén R.
Life Sciences scimago Q1 wos Q1 Open Access
2021-03-01 citations by CoLab: 32 Abstract  
Aims Breast cancer is one of the leading causes of woman deaths worldwide, being a major public health problem. It has been reported that the expression of the RNA-editing enzyme Adenosine Deaminase Acting on RNAs 1 (ADAR1) is upregulated in breast cancer, predicting poor prognosis in patients. A few reports in literature examine ADAR1 and long non-coding RNAs (lncRNAs) interplay in cancer and suggest key roles in cancer-related pathways. This study aimed to investigate whether ADAR1 could alter the expression levels of lncRNAs and explore how those changes are related to breast cancer biology. Main methods ADAR1 overexpression and knockdown studies were performed in breast cancer cell lines to analyze the effects over lncRNAs expression. Guilt-by-Association correlation analysis of the TCGA-BRCA cohort was performed to predict the function of the lncRNA LINC00944. Key findings Here, we show that LINC00944 is responsive to ADAR1 up- and downregulation in breast cancer cells. We found that LINC00944 expression has a strong relationship with immune signaling pathways. Further assessment of the TCGA-BRCA cohort showed that LINC00944 expression was positively correlated to tumor-infiltrating T lymphocytes and pro-apoptotic markers. Moreover, we found that LINC00944 expression was correlated to the age at diagnosis, tumor size, and estrogen and progesterone receptor expression. Finally, we show that low expression of LINC00944 is correlated to poor prognosis in breast cancer patients. Significance Our study provides further evidence of the effect of ADAR1 over lncRNA expression levels, and on the participation of LINC00944 in breast cancer, suggesting to further investigate its potential role as prognostic biomarker.
Garretto A., Miller-Ensminger T., Ene A., Merchant Z., Shah A., Gerodias A., Biancofiori A., Canchola S., Canchola S., Castillo E., Chowdhury T., Gandhi N., Hamilton S., Hatton K., Hyder S., et. al.
Frontiers in Microbiology scimago Q1 wos Q2 Open Access
2020-09-04 citations by CoLab: 42 PDF Abstract  
Urinary tract infections (UTIs) are one of the most common human bacterial infections. While UTIs are commonly associated with colonization by Escherichia coli, members of this species also have been found within the bladder of individuals with no lower urinary tract symptoms, also known as asymptomatic bacteriuria. Prior studies have found that both uropathogenic E. coli (UPEC) strains and E. coli isolates that are not associated with UTIs encode for virulence factors. Thus, the reason(s) why E. coli sometimes causes UTI-like symptoms remain(s) elusive. In this study, the genomes of 66 E. coli isolates from adult female bladders were sequenced. These isolates were collected from four cohorts, including women: (1) without lower urinary tract symptoms, (2) overactive bladder symptoms, (3) urgency urinary incontinence, and (4) a clinical diagnosis of UTI. Comparative genomic analyses were conducted, including core and accessory genome analyses, virulence and motility gene analyses, and antibiotic resistance prediction and testing. We found that the genomic content of these 66 E. coli isolates does not correspond with the participant’s symptom status. We thus looked beyond the E. coli genomes to the composition of the entire urobiome and found that the presence of E. coli alone was not sufficient to distinguish between the urobiomes of individuals with UTI and those with no lower urinary tract symptoms. Because E. coli presence, abundance, and genomic content appear to be weak predictors of UTI status, we hypothesize that UTI symptoms associated with detection of E. coli are more likely the result of urobiome composition.
Magruder M., Edusei E., Zhang L., Albakry S., Satlin M.J., Westblade L.F., Malha L., Sze C., Lubetzky M., Dadhania D.M., Lee J.R.
Gut Microbes scimago Q1 wos Q1 Open Access
2020-08-30 citations by CoLab: 59 PDF
Takahashi D., Hoshina N., Kabumoto Y., Maeda Y., Suzuki A., Tanabe H., Isobe J., Yamada T., Muroi K., Yanagisawa Y., Nakamura A., Fujimura Y., Saeki A., Ueda M., Matsumoto R., et. al.
EBioMedicine scimago Q1 wos Q1 Open Access
2020-08-01 citations by CoLab: 115 Abstract  
Background Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disorder with a high prevalence, especially in industrialized countries. Dysbiosis of the intestinal microbiota has been observed in RA patients. For instance, new-onset untreated RA (NORA) is associated with the underrepresentation of the Clostridium cluster XIVa, including Lachnospiraceae, which are major butyrate producers, although the pathological relevance has remained obscure. Follicular regulatory T (TFR) cells play critical regulatory roles in the pathogenesis of autoimmune diseases, including RA. Reduced number of circulating TFR cells has been associated with the elevation of autoantibodies and disease severity in RA. However, the contribution of commensal microbe-derived butyrate in controlling TFR cell differentiation remains unknown. Methods We examined the contribution of microbe-derived butyrate in controlling autoimmune arthritis using collagen-induced arthritis (CIA) and SKG arthritis models. We phenotyped autoimmune responses in the gut-associated lymphoid tissues (GALT) in the colon and joint-draining lymph nodes in the CIA model. We developed an in vitro CXCR5+Bcl-6+Foxp3+ TFR (iTFR) cell culture system and examined whether butyrate promotes the differentiation of iTFR cells. Findings Microbe-derived butyrate suppressed the development of autoimmune arthritis. The immunization of type II collagen (CII) caused hypertrophy of the GALT in the colon by amplifying the GC reaction prior to the onset of the CIA. Butyrate mitigated these pathological events by promoting TFR cell differentiation. Butyrate directly induced the differentiation of functional TFR cells in vitro by enhancing histone acetylation in TFR cell marker genes. This effect was attributed to histone deacetylase (HDAC) inhibition by butyrate, leading to histone hyperacetylation in the promoter region of the TFR-cell marker genes. The adoptive transfer of the butyrate-treated iTFR cells reduced CII-specific autoantibody production and thus ameliorated the symptoms of arthritis. Interpretation Accordingly, microbiota-derived butyrate serves as an environmental cue to enhance TFR cells, which suppress autoantibody production in the systemic lymphoid tissue, eventually ameliorating RA. Our findings provide mechanistic insights into the link between the gut environment and RA risk. Funding This work was supported by AMED-Crest (16gm1010004h0101, 17gm1010004h0102, 18gm1010004h0103, and 19gm1010004s0104 to KH), the Japan Society for the Promotion of Science (JP17KT0055, JP16H01369, and JP18H04680 to KH; JP17K15734 to DT), Keio University Special Grant-in-Aid for Innovative Collaborative Research Projects (KH), Keio Gijuku Fukuzawa Memorial Fund for the Advancement of Education and Research (DT), the SECOM Science and Technology Foundation (KH), the Cell Science Research Foundation (KH), the Mochida Memorial Foundation for Medical and Pharmaceutical Research (DT), the Suzuken Memorial Foundation (KH and DT), the Takeda Science Foundation (KH and DT), The Science Research Promotion Fund, and The Promotion and Mutual Aid Corporation for Private Schools of Japan (KH).
Zhang S., Dogan B., Guo C., Herlekar D., Stewart K., Scherl E.J., Simpson K.W.
Antibiotics scimago Q1 wos Q1 Open Access
2020-07-30 citations by CoLab: 52 PDF Abstract  
Short chain fatty acids (SCFA), principally acetate, propionate, and butyrate, are produced by fermentation of dietary fibers by the gut microbiota. SCFA regulate the growth and virulence of enteric pathogens, such as enterohemorrhagic E. coli (EHEC), Klebsiella and Salmonella. We sought to investigate the impact of SCFA on growth and virulence of pathosymbiont E. coli associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC), and their role in regulating host responses to bacterial infection in vitro. We found that under ileal conditions (pH = 7.4; 12 mM total SCFA), SCFA significantly (p < 0.05) potentiate the growth and motility of pathosymbiont E. coli. However, under colonic conditions (pH = 6.5; 65 to 123 mM total SCFA), SCFA significantly (p < 0.05) inhibit growth in a pH dependent fashion (up to 60%), and down-regulate virulence gene expression (e.g., fliC, fimH, htrA, chuA, pks). Functional analysis reveals that colonic SCFA significantly (p < 0.05) inhibit E. coli motility (up to 95%), infectivity (up to 60%), and type 1 fimbria-mediated agglutination (up to 50%). In addition, SCFA significantly (p < 0.05) inhibit the activation of NF-kB, and IL-8 production by epithelial cells. Our findings provide novel insights on the role of the regional chemical microenvironment in regulating the growth and virulence of pathosymbiont E. coli and opportunities for therapeutic intervention.
Rosser E.C., Piper C.J., Matei D.E., Blair P.A., Rendeiro A.F., Orford M., Alber D.G., Krausgruber T., Catalan D., Klein N., Manson J.J., Drozdov I., Bock C., Wedderburn L.R., Eaton S., et. al.
Cell Metabolism scimago Q1 wos Q1
2020-04-01 citations by CoLab: 379 Abstract  
The differentiation of IL-10-producing regulatory B cells (Bregs) in response to gut-microbiota-derived signals supports the maintenance of tolerance. However, whether microbiota-derived metabolites can modulate Breg suppressive function remains unknown. Here, we demonstrate that rheumatoid arthritis (RA) patients and arthritic mice have a reduction in microbial-derived short-chain fatty acids (SCFAs) compared to healthy controls and that in mice, supplementation with the SCFA butyrate reduces arthritis severity. Butyrate supplementation suppresses arthritis in a Breg-dependent manner by increasing the level of the serotonin-derived metabolite 5-Hydroxyindole-3-acetic acid (5-HIAA), which activates the aryl-hydrocarbon receptor (AhR), a newly discovered transcriptional marker for Breg function. Thus, butyrate supplementation via AhR activation controls a molecular program that supports Breg function while inhibiting germinal center (GC) B cell and plasmablast differentiation. Our study demonstrates that butyrate supplementation may serve as a viable therapy for the amelioration of systemic autoimmune disorders.
Magruder M., Sholi A.N., Gong C., Zhang L., Edusei E., Huang J., Albakry S., Satlin M.J., Westblade L.F., Crawford C., Dadhania D.M., Lubetzky M., Taur Y., Littman E., Ling L., et. al.
Nature Communications scimago Q1 wos Q1 Open Access
2019-12-04 citations by CoLab: 160 PDF Abstract  
The origin of most bacterial infections in the urinary tract is often presumed to be the gut. Herein, we investigate the relationship between the gut microbiota and future development of bacteriuria and urinary tract infection (UTI). We perform gut microbial profiling using 16S rRNA gene deep sequencing on 510 fecal specimens from 168 kidney transplant recipients and metagenomic sequencing on a subset of fecal specimens and urine supernatant specimens. We report that a 1% relative gut abundance of Escherichia is an independent risk factor for Escherichia bacteriuria and UTI and a 1% relative gut abundance of Enterococcus is an independent risk factor for Enterococcus bacteriuria. Strain analysis establishes a close strain level alignment between species found in the gut and in the urine in the same subjects. Our results support a gut microbiota–UTI axis, suggesting that modulating the gut microbiota may be a potential novel strategy to prevent UTIs. Urinary tract infections (UTIs) are associated with changes in the gut microbiome. Here, the authors evaluate the relationship between the gut microbiome and development of UTI in kidney transplant patients and show that uropathogenic gut abundance might represent a risk factor for development of bacteriuria and UTI.
Cryan J.F., O'Riordan K.J., Cowan C.S., Sandhu K.V., Bastiaanssen T.F., Boehme M., Codagnone M.G., Cussotto S., Fulling C., Golubeva A.V., Guzzetta K.E., Jaggar M., Long-Smith C.M., Lyte J.M., Martin J.A., et. al.
Physiological Reviews scimago Q1 wos Q1
2019-10-01 citations by CoLab: 3007 Abstract  
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Thänert R., Reske K.A., Hink T., Wallace M.A., Wang B., Schwartz D.J., Seiler S., Cass C., Burnham C.D., Dubberke E.R., Kwon J.H., Dantas G.
mBio scimago Q1 wos Q1 Open Access
2019-08-27 citations by CoLab: 79 PDF Abstract  
The increasing antimicrobial resistance of uropathogens is challenging the continued efficacy of empiric antibiotic therapy for UTIs, which are among the most frequent bacterial infections worldwide. It has been suggested that drug-resistant uropathogens could persist in the intestine after the resolution of UTI and cause recurrences following periurethral contamination. A better understanding of the transmission dynamics between the intestinal and urinary tracts, combined with phenotypic characterization of the uropathogen populations in both habitats, could inform prudent therapies designed to overcome the rising resistance of uropathogens. Here, we integrate genomic surveillance with clinical microbiology to show that drug-resistant clones persist within and are readily transmitted between the intestinal and urinary tracts of patients affected by recurrent and nonrecurrent UTIs. Thus, our results advocate for understanding persistent intestinal uropathogen colonization as part of the pathophysiology of UTIs, particularly in patients affected by recurrent episodes of symptomatic disease.
Forde B.M., Roberts L.W., Phan M., Peters K.M., Fleming B.A., Russell C.W., Lenherr S.M., Myers J.B., Barker A.P., Fisher M.A., Chong T., Yin W., Chan K., Schembri M.A., Mulvey M.A., et. al.
Nature Communications scimago Q1 wos Q1 Open Access
2019-08-13 citations by CoLab: 92 PDF Abstract  
Recurrent urinary tract infections (rUTIs) are extremely common, with ~ 25% of all women experiencing a recurrence within 1 year of their original infection. Escherichia coli ST131 is a globally dominant multidrug resistant clone associated with high rates of rUTI. Here, we show the dynamics of an ST131 population over a 5-year period from one elderly woman with rUTI since the 1970s. Using whole genome sequencing, we identify an indigenous clonal lineage (P1A) linked to rUTI and persistence in the fecal flora, providing compelling evidence of an intestinal reservoir of rUTI. We also show that the P1A lineage possesses substantial plasmid diversity, resulting in the coexistence of antibiotic resistant and sensitive intestinal isolates despite frequent treatment. Our longitudinal study provides a unique comprehensive genomic analysis of a clonal lineage within a single individual and suggests a population-wide resistance mechanism enabling rapid adaptation to fluctuating antibiotic exposure. Recurrent urinary tract infections occur in ~ 25% of women. Here, Beatson and colleagues use whole genome sequencing to track the dynamics of an E. coli ST131 clone in a single patient over a 5-year period. This study provides unique insights into pathogen evolution during recurrent urinary infection.
Coburn B., Science M., Lee K., Tomlinson C., Bansal S., Brown K.A., Timberlake K., Rooney A.M.
Clinical Infectious Diseases scimago Q1 wos Q1
2019-08-01 citations by CoLab: 33 Abstract  
AbstractBackgroundDiscontinuation of inappropriate antimicrobial therapy is an important target for stewardship intervention. The drug and duration-dependent effects of antibiotics on the developing neonatal gut microbiota needs to be precisely quantified.MethodsIn this retrospective, cross-sectional study, we performed 16S rRNA sequencing on stool swab samples collected from neonatal intensive care unit patients within 7 days of discontinuation of therapy who received ampicillin and tobramycin (AT), ampicillin and cefotaxime (AC), or ampicillin, tobramycin, and metronidazole (ATM). We compared taxonomic composition within term and preterm infant groups between treatment regimens. We calculated adjusted effect estimates for antibiotic type and duration of therapy on the richness of obligate anaerobes and known butyrate-producers in all infants.ResultsA total of 72 infants were included in the study. Term infants received AT (20/28; 71%) or AC (8/28; 29%) with median durations of 3 and 3.5 days, respectively. Preterm infants received AT (32/44; 73%) or ATM (12/44; 27%) with median durations of 4 and 7 days, respectively. Compositional analyses of 67 stool swab samples demonstrated low diversity and dominance by potential pathogens. Within 1 week of discontinuation of therapy, each additional day of antibiotics was associated with lower richness of obligate anaerobes (adjusted risk ratio [aRR], 0.84; 95% confidence interval [CI], .73–.95) and butyrate-producers (aRR, 0.82; 95% CI, .67–.97).ConclusionsEach additional day of antibiotics was associated with lower richness of anaerobes and butyrate-producers within 1 week after therapy. A longitudinally sampled cohort with preexposure sampling is needed to validate our results.
Dang A.T., Marsland B.J.
Mucosal Immunology scimago Q1 wos Q1
2019-07-01 citations by CoLab: 669 Abstract  
The microbiota plays an essential role in the education, development, and function of the immune system, both locally and systemically. Emerging experimental and epidemiological evidence highlights a crucial cross-talk between the intestinal microbiota and the lungs, termed the ‘gut–lung axis’. Changes in the constituents of the gut microbiome, through either diet, disease or medical interventions (such as antibiotics) is linked with altered immune responses and homeostasis in the airways. The importance of the gut–lung axis has become more evident following the identification of several gut microbe-derived components and metabolites, such as short-chain fatty acids (SCFAs), as key mediators for setting the tone of the immune system. Recent studies have supported a role for SCFAs in influencing hematopoietic precursors in the bone marrow—a major site of innate and adaptive immune cell development. Here, we review the current understanding of host–microbe cross-talk along the gut–lung axis. We highlight the importance of SCFAs in shaping and promoting bone marrow hematopoiesis to resolve airway inflammation and to support a healthy homeostasis.
Zhang J., Lei Y., Du H., Li Z., Wang X., Yang D., Gao F., Li J.
2025-04-01 citations by CoLab: 0 PDF Abstract  
The traditional view of sterile urine has been challenged by advancements in next-generation sequencing, revealing that the urinary microbiome significantly influences individual health and various urinary system diseases. Urinary tract infections in patients with neurogenic bladder are highly prevalent, recurrent, and lifelong. If frequent urinary tract infections are not adequately managed, they may ultimately lead to chronic renal failure. The excessive use of antibiotics to prevent and treat urinary tract infections may lead to increased bacterial resistance, limiting future therapeutic options. This review summarizes commonly used microbiome research techniques and urine collection methods, compiles current studies on the urinary microbiome in neurogenic bladder patients, and discusses the potential implications of urinary microbiome composition for preventing, diagnosing, and treating urinary tract infections. By summarizing current research findings, we aim to enhance understanding of the urinary microbiome in neurogenic bladder patients and promote the standardization and clinical translation of microbiome research.
DeVeaux A.L., Hall-Moore C., Shaikh N., Wallace M., Burnham C.D., Schnadower D., Kuppermann N., Mahajan P., Ramilo O., Tarr P.I., Dantas G., Schwartz D.J.
Microbiome scimago Q1 wos Q1 Open Access
2025-03-24 citations by CoLab: 0 PDF
Moreland R.B., Brubaker L., Tinawi L., Wolfe A.J.
Clinical Microbiology Reviews scimago Q1 wos Q1
2025-03-13 citations by CoLab: 2 Abstract  
SUMMARY Urinary tract infection (UTI) is among the most common infections in clinical practice. In some cases, if left untreated, it can lead to pyelonephritis and urosepsis. In other cases, UTI resolves without treatment. Clinical diagnosis is typically based on patient symptoms and/or urinalysis, including urine dipsticks. The standard urine culture method is sometimes employed to identify the suspected urinary pathogen (uropathogen) and/or guide antimicrobial choice, but results are rarely available before 24 h. The standard urine culture method also misses fastidious, anaerobic, and slow-growing uropathogens and rarely reports polymicrobial infections. The unexplained combination of negative urine cultures with persistent urinary tract symptoms is distressing to both patients and clinicians. Given the broad appreciation of the advantages provided by rapid testing (e.g., for COVID-19 or influenza A), a rapid, accurate diagnostic test is needed to deliver timely treatment to patients seeking care for UTI that optimizes antibiotic stewardship. Herein, we discuss progress being made toward an accessible, timely (i.e., within hours), accurate assay with results that are clinically useful for the treating clinician within the timeframe of the infection (i.e., the growth rate of the pathogen(s)). New and emerging uropathogens often overlooked by current diagnostic techniques are also reviewed.
Azimzadeh P.N., Birchenough G.M., Gualbuerto N.C., Pinkner J.S., Tamadonfar K.O., Beatty W., Hannan T.J., Dodson K.W., Ibarra E.C., Kim S., Schreiber H.L., Janetka J.W., Kau A.L., Earl A.M., Miller M.J., et. al.
Science advances scimago Q1 wos Q1 Open Access
2025-01-31 citations by CoLab: 0 PDF Abstract  
Urinary tract infections (UTIs) are highly recurrent and frequently caused by Uropathogenic Escherichia coli (UPEC) strains that can be found in patient intestines. Seeding of the urinary tract from this intestinal reservoir likely contributes to UTI recurrence (rUTI) rates. Thus, understanding the factors that promote UPEC intestinal colonization is of critical importance to designing therapeutics to reduce rUTI incidence. Although E. coli is found in high abundance in large intestine mucus, little is known about how it is able to maintain residence in this continuously secreted hydrogel. We discovered that the FimH adhesin of type 1 pili (T1P) bound throughout the secreted mucus layers of the colon and to epithelial cells in mouse and human samples. Disruption of T1P led to reduced association with colon mucus. Notably, this mutant up-regulated flagellar production and infiltrated the protective inner mucus layer of the colon. This could explain how UPEC resists being washed off by the continuously secreted mucus layers of the colon.
Kacena C.
Nutrition Reviews scimago Q1 wos Q1
2025-01-28 citations by CoLab: 0 Abstract  
Abstract The gut microbiome is a complex system that directly interacts with and influences many systems in the body. This delicate balance of microbiota plays an important role in health and disease and is highly influenced by lifestyle factors and the surrounding environment. As further research emerges, understanding the full potential of the gut microbiome and the impact of using nutraceuticals to positively influence its function may open the door to greater therapeutic outcomes in the treatment and prevention of disease. Curcumin, a bioactive compound derived from the turmeric rhizome, has been studied in depth for its influence on human health as a potent anti-inflammatory and antioxidant properties. However, the therapeutic activity of curcumin is limited by its low oral bioavailability. While most available research has primarily focused on the curcuminoid compounds of turmeric, the non-curcuminoid compounds hold promise to offer therapeutic benefits while synergistically enhancing the bioavailability of curcumin and supporting the gut microbiome. This review summarizes current knowledge of the relationship between the gut and the various systems within the body, and how dysbiosis, or disruption in the gut microbial balance, leads to inflammation and increased risk of chronic disease. The review also summarizes recent research that focuses on the bioactivity of both the curcuminoid and non-curcuminoid compounds that comprise the whole turmeric root and their synergistic role in enhancing bioavailability to support a healthy gut microbiome and promising use in the treatment and prevention of disease.
Krohmals S., de Terwangne C., Devresse A., Goffin E., Darius T., Buemi A., Mourad M., Rodriguez-Villalobos H., Kanaan N.
Journal of Clinical Medicine scimago Q1 wos Q1 Open Access
2025-01-18 citations by CoLab: 0 PDF Abstract  
Background: Urinary tract infections (UTIs) are a common complication after kidney transplantation. The aim of this study was to evaluate the impact of pre-existing diabetes mellitus and post-transplant diabetes mellitus (PTDM) on the occurrence of pyelonephritis in kidney transplant recipients. Methods: We performed a retrospective analysis which included 299 adult patients transplanted with a kidney between 2018 and 2022. Patients were categorized into pre-transplantation diabetics, PTDM, and non-diabetics. Asymptomatic bacteriuria and lower urinary infections were not included. Results: During a median follow-up time of 31 [17–45] months, 100 UTIs were reported in the total cohort, with a mean time from transplantation to the first UTI episode of 10 ± 11 months. At 48 months, the cumulative incidence of UTIs was 34.9%, 56%, and 47.3% for patients without prior diabetes, pre-transplant diabetes, and PTDM, respectively. Pre-transplant diabetes was independently associated with 79% increased risk of UTIs (adjusted HR = 1.79, 95% CI = 1.14–2.81, p = 0.011). The risk associated with female gender increased to 85%. Patient survival was not significantly affected by the interaction between diabetes and UTI occurrence. Conclusions: Pre-transplant diabetes arises as a significant risk factor for UTIs after kidney transplantation.
Chen L., Xu Q., Chen W., Liu J., Xu T., Yang J., Ji L.
2025-01-03 citations by CoLab: 0 Abstract  
Limited research into the tumor immune microenvironment (TIME) for bladder urothelial carcinoma (BUC), particularly the neglect of the intratumoral microbiota, has hindered the development of immunotherapies targeting BUC. Here, we collect 401 patients with BUC with host transcriptome samples and matched tumor microbiome samples from The Cancer Genome Atlas database. Besides, two independent BUC cohorts receiving immunotherapy were obtained. First, we find that the TIME profile is closely related to the prognosis of patients with BUC. Additionally, the genus Lachnoclostridium in tumors could regulate the accumulation of chemokines to recruit immune cell populations into bladder tumors. Among them, chemokines include CCL3, CCL4, CXCL9, CXCL10, and CXCL11, and immune cells mainly involve macrophages and CD8+ T cells. Analyses based on two independent immunotherapy cohorts suggest that these immune-related chemokines strongly influence the immunotherapeutic efficacy of BUC. Furthermore, drug predictive analyses show that immune-related chemokines impact patients' sensitivity to diverse drugs. These results suggest a dual role of immune-related chemokines in combination therapy against BUC. Collectively, our study provides new insights into the regulation of TIME by intratumoral microbiota and provides guidance for improving immunotherapy against BUC.
Guo C., Tang Q., Yuan J., Li S., Yang X., Li Y., Zhou X., Ji H., Qin Y., Wu L.
Talanta scimago Q1 wos Q1
2025-01-01 citations by CoLab: 0 Abstract  
The accurate discrimination of bacterial infection is imperative for precise clinical diagnosis and treatment. Here, this work presents a simplified sensor array utilizing "All-in-One" Pdots for efficient discrimination of diverse bacterial samples. The "All-in-One" Pdots sensor (AOPS) were synthesized using three components that exhibit fluorescence resonance energy transfer (FRET) effect, facilitating the efficient integration of multiple discrimination channels to generate specific fluorescence response patterns through a single detection under single-wavelength excitation. Additionally, machine learning techniques were employed to visually represent the fluorescence response patterns of AOPS upon exposure to bacterial metabolites derived from diverse bacterial species. The as-prepared sensor platform demonstrated excellent performance in analyzing eight common bacteria, drug-resistant strains, mixed bacterial samples, bacterial biofilms and real samples, presenting significant potential in the identification of complex samples for bacterial analysis.
Vautrin N., Dahyot S., Leoz M., Caron F., Grand M., Feldmann A., Gravey F., Legris S., Ribet D., Alexandre K., Pestel-Caron M.
Virulence scimago Q1 wos Q1 Open Access
2024-12-26 citations by CoLab: 0 PDF
Lv Y., Xian Y., Lei X., Xie S., Zhang B.
Medicine (United States) scimago Q3 wos Q2 Open Access
2024-12-13 citations by CoLab: 1 Abstract  
Pediatric obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder associated with significant neurocognitive and behavioral impairments. Recent studies have highlighted the role of gut microbiota and the microbiota-gut-brain axis (MGBA) in influencing cognitive health in children with OSA. This narrative review aims to summarize current knowledge on the relationship between gut microbiota, MGBA, and cognitive function in pediatric OSA. It also explores the potential of artificial intelligence and machine learning in advancing this field and identifying novel therapeutic strategies. Pediatric OSA is associated with gut dysbiosis, reduced microbial diversity, and metabolic disruptions. MGBA mechanisms, such as endocrine, immune, and neural pathways, link gut microbiota to cognitive outcomes. Artificial intelligence and machine learning methodologies offer promising tools to uncover microbial markers and mechanisms associated with cognitive deficits in OSA. Future research should focus on validating these findings through clinical trials and developing personalized therapeutic approaches targeting the gut microbiota.
Zou J., Xu B., Gao H., Luo P., Chen T., Duan H.
Frontiers in Immunology scimago Q1 wos Q1 Open Access
2024-12-05 citations by CoLab: 0 PDF Abstract  
Urological tumors are an important disease affecting global human health, and their pathogenesis and treatment have been the focus of medical research. With the in - depth study of microbiomics, the role of the microbiome in urological tumors has gradually attracted attention. However, the current research on tumor - associated microorganisms mostly focuses on one type or one site, and currently, there is a lack of attention to the microbiome in the immunity and immunotherapy of urological tumors. Therefore, in this paper, we systematically review the distribution characteristics of the microbiome (including microorganisms in the gut, urine, and tumor tissues) in urologic tumors, the relationship with disease prognosis, and the potential mechanisms of microbial roles in immunotherapy. In particular, we focus on the molecular mechanisms by which the microbiome at different sites influences tumor immunity through multiple “messengers” and pathways. We aim to further deepen the understanding of microbiome mechanisms in urologic tumors, and also point out the direction for the future development of immunotherapy for urologic tumors.

Top-30

Journals

1
2
3
1
2
3

Publishers

5
10
15
20
25
5
10
15
20
25
  • We do not take into account publications without a DOI.
  • Statistics recalculated only for publications connected to researchers, organizations and labs registered on the platform.
  • Statistics recalculated weekly.

Are you a researcher?

Create a profile to get free access to personal recommendations for colleagues and new articles.
Share
Cite this
GOST | RIS | BibTex | MLA
Found error?