48 results on '"Petrosino, Joseph F."'
Search Results
2. Human milk oligosaccharide DSLNT and gut microbiome in preterm infants predicts necrotising enterocolitis
- Author
-
Masi, Andrea C, Embleton, Nicholas D, Lamb, Christopher A, Young, Gregory, Granger, Claire L, Najera, Julia, Smith, Daniel P, Hoffman, Kristi L, Petrosino, Joseph F, Bode, Lars, Berrington, Janet E, and Stewart, Christopher J
- Abstract
ObjectiveNecrotising enterocolitis (NEC) is a devastating intestinal disease primarily affecting preterm infants. The underlying mechanisms are poorly understood: mother’s own breast milk (MOM) is protective, possibly relating to human milk oligosaccharide (HMO) and infant gut microbiome interplay. We investigated the interaction between HMO profiles and infant gut microbiome development and its association with NEC.DesignWe performed HMO profiling of MOM in a large cohort of infants with NEC (n=33) with matched controls (n=37). In a subset of 48 infants (14 with NEC), we also performed longitudinal metagenomic sequencing of infant stool (n=644).ResultsConcentration of a single HMO, disialyllacto-N-tetraose (DSLNT), was significantly lower in MOM received by infants with NEC compared with controls. A MOM threshold level of 241 nmol/mL had a sensitivity and specificity of 0.9 for NEC. Metagenomic sequencing before NEC onset showed significantly lower relative abundance of Bifidobacterium longumand higher relative abundance of Enterobacter cloacaein infants with NEC. Longitudinal development of the microbiome was also impacted by low MOM DSLNT associated with reduced transition into preterm gut community types dominated by Bifidobacteriumspp and typically observed in older infants. Random forest analysis combining HMO and metagenome data before disease accurately classified 87.5% of infants as healthy or having NEC.ConclusionThese results demonstrate the importance of HMOs and gut microbiome in preterm infant health and disease. The findings offer potential targets for biomarker development, disease risk stratification and novel avenues for supplements that may prevent life-threatening disease.
- Published
- 2021
- Full Text
- View/download PDF
3. Rotavirus infection induces glycan availability to promote ileum-specific changes in the microbiome aiding rotavirus virulence
- Author
-
Engevik, Melinda A., Banks, Lori D., Engevik, Kristen A., Chang-Graham, Alexandra L., Perry, Jacob L., Hutchinson, Diane S., Ajami, Nadim J., Petrosino, Joseph F., and Hyser, Joseph M.
- Abstract
ABSTRACTMultiple studies have identified changes within the gut microbiome in response to diarrheal-inducing bacterial pathogens. However, examination of the microbiome in response to viral pathogens remains understudied. Compounding this, many studies use fecal samples to assess microbiome composition; which may not accurately mirror changes within the small intestine, the primary site for most enteric virus infections. As a result, the functional significance of small intestinal microbiome shifts during infection is not well defined. To address these gaps, rotavirus-infected neonatal mice were examined for changes in bacterial community dynamics, host gene expression, and tissue recovery during infection. Profiling bacterial communities using 16S rRNA sequencing suggested significant and distinct changes in ileal communities in response to rotavirus infection, with no significant changes for other gastrointestinal (GI) compartments. At 1-d post-infection, we observed a loss in Lactobacillusspecies from the ileum, but an increase in Bacteroidesand Akkermansia, both of which exhibit mucin-digesting capabilities. Concomitant with the bacterial community shifts, we observed a loss of mucin-filled goblet cells in the small intestine at d 1, with recovery occurring by d 3. Rotavirus infection of mucin-producing cell lines and human intestinal enteroids (HIEs) stimulated release of stored mucin granules, similar to in vivofindings. In vitro, incubation of mucins with Bacteroidesor Akkermansiamembers resulted in significant glycan degradation, which altered the binding capacity of rotavirus in silicoand in vitro. Taken together, these data suggest that the response to and recovery from rotavirus-diarrhea is unique between sub-compartments of the GI tract and may be influenced by mucin-degrading microbes.
- Published
- 2020
- Full Text
- View/download PDF
4. The Fecal Microbiome in Infants With Biliary Atresia Associates With Bile Flow After Kasai Portoenterostomy
- Author
-
Tessier, Mary Elizabeth M., Cavallo, Laurel, Yeh, Jennifer, Harpavat, Sanjiv, Hoffman, Kristi L., Petrosino, Joseph F., and Shneider, Benjamin L.
- Abstract
Biliary atresia's (BA) response to surgical Kasai portoenterostomy (KP) is uneven and dependent upon bile flow; 50% of infants require a liver transplant by 24 months. We hypothesized that the microbiome may identify and associate with outcomes in BA. Stool samples were collected from infants with cholestasis (n?=?15), 8 of which with BA were followed longitudinally.16S sequencing was performed on all samples (n?=?45). Whole Genome Sequencing (WGS) was performed on BA pre-KP samples (n?=?8). Infants with BA, other forms of cholestasis, BA infants with very good bile flow (VGBF) and not (nVGBF) (VGBF dichotomized by TSBA <40?µmol/L by 6 months) were compared. Of the 8 infants with BA, 4 infants had VGBF. Microbial richness was inversely proportional to degree of cholestasis (P?=?0.046). Increased Bifidobacteriumabundance associated with VGBF (P?=?0.03) and decreased cholestasis (P?0.01) at 1 month post-KP. Pre-KP, community structure differed in infants with BA versus other cholestasis. Interestingly, infants who subsequently achieved VGBF had increased diversity (P?=?0.03) and different community structure at the pre-KP time point. WGS corroborated Bifidobacterium'spre-KP importance. The microbiome differs between infants with BA and other cholestasis. It additionally differs between infants with BA who have good and poor bile flow, and thus outcomes, post-KP. These differences are seen even before KP. These data suggest that bile influences the development of the infant microbiome and that there may be possible influences of the pre- and post-KP microbiome on bile flow after KP. Further larger studies are needed to confirm these findings.
- Published
- 2020
- Full Text
- View/download PDF
5. Metagenomics of the faecal virome indicate a cumulative effect of enterovirus and gluten amount on the risk of coeliac disease autoimmunity in genetically at risk children: the TEDDY study
- Author
-
Lindfors, Katri, Lin, Jake, Lee, Hye-Seung, Hyo¨ty, Heikki, Nykter, Matti, Kurppa, Kalle, Liu, Edwin, Koletzko, Sibylle, Rewers, Marian, Hagopian, William, Toppari, Jorma, Ziegler, Annette-Gabriele, Akolkar, Beena, Krischer, Jeffrey P, Petrosino, Joseph F, Lloyd, Richard E, and Agardh, Daniel
- Abstract
ObjectiveHigher gluten intake, frequent gastrointestinal infections and adenovirus, enterovirus, rotavirus and reovirus have been proposed as environmental triggers for coeliac disease. However, it is not known whether an interaction exists between the ingested gluten amount and viral exposures in the development of coeliac disease. This study investigated whether distinct viral exposures alone or together with gluten increase the risk of coeliac disease autoimmunity (CDA) in genetically predisposed children.DesignThe Environmental Determinants of Diabetes in the Young study prospectively followed children carrying the HLA risk haplotypes DQ2 and/or DQ8 and constructed a nested case–control design. From this design, 83 CDA case–control pairs were identified. Median age of CDA was 31 months. Stool samples collected monthly up to the age of 2 years were analysed for virome composition by Illumina next-generation sequencing followed by comprehensive computational virus profiling.ResultsThe cumulative number of stool enteroviral exposures between 1 and 2 years of age was associated with an increased risk for CDA. In addition, there was a significant interaction between cumulative stool enteroviral exposures and gluten consumption. The risk conferred by stool enteroviruses was increased in cases reporting higher gluten intake.ConclusionsFrequent exposure to enterovirus between 1 and 2 years of age was associated with increased risk of CDA. The increased risk conferred by the interaction between enteroviruses and higher gluten intake indicate a cumulative effect of these factors in the development of CDA.
- Published
- 2020
- Full Text
- View/download PDF
6. Prospective virome analyses in young children at increased genetic risk for type 1 diabetes
- Author
-
Vehik, Kendra, Lynch, Kristian F., Wong, Matthew C., Tian, Xiangjun, Ross, Matthew C., Gibbs, Richard A., Ajami, Nadim J., Petrosino, Joseph F., Rewers, Marian, Toppari, Jorma, Ziegler, Anette G., She, Jin-Xiong, Lernmark, Ake, Akolkar, Beena, Hagopian, William A., Schatz, Desmond A., Krischer, Jeffrey P., Hyöty, Heikki, and Lloyd, Richard E.
- Abstract
Viruses are implicated in autoimmune destruction of pancreatic islet β cells, which results in insulin deficiency and type 1 diabetes (T1D)1–4. Certain enteroviruses can infect β cells in vitro5, have been detected in the pancreatic islets of patients with T1D6and have shown an association with T1D in meta-analyses4. However, establishing consistency in findings across studies has proven difficult. Obstacles to convincingly linking RNA viruses to islet autoimmunity may be attributed to rapid viral mutation rates, the cyclical periodicity of viruses7and the selection of variants with altered pathogenicity and ability to spread in populations. β cells strongly express cell-surface coxsackie and adenovirus receptor (CXADR) genes, which can facilitate enterovirus infection8. Studies of human pancreata and cultured islets have shown significant variation in enteroviral virulence to β cells between serotypes and within the same serotype9,10. In this large-scale study of known eukaryotic DNA and RNA viruses in stools from children, we evaluated fecally shed viruses in relation to islet autoimmunity and T1D. This study showed that prolonged enterovirus B rather than independent, short-duration enterovirus B infections may be involved in the development of islet autoimmunity, but not T1D, in some young children. Furthermore, we found that fewer early-life human mastadenovirus C infections, as well as CXADRrs6517774, independently correlated with islet autoimmunity.
- Published
- 2019
- Full Text
- View/download PDF
7. Prebiotics, Probiotics, and Acetate Supplementation Prevent Hypertension in a Model of Obstructive Sleep Apnea.
- Author
-
Ganesh, Bhanu P., Nelson, James W., Ganesan, Arunkumar, Bryan, Robert M., Durgan, David J., Eskew, Joshua R., Ajami, Nadim J., Petrosino, Joseph F., and Bryan, Robert M Jr
- Abstract
Disruption of the gut microbiota, termed gut dysbiosis, has been described in animal models of hypertension and hypertensive patients. We have shown that gut dysbiosis plays a causal role in the development of hypertension in a rat model of obstructive sleep apnea (OSA). Functional analysis of the dysbiotic microbiota in OSA demonstrates a loss of short chain fatty acid-producing bacteria. However, measurements of short chain fatty acid concentrations and testing of their role in blood pressure regulation are lacking. We hypothesized that reduced short chain fatty acids in the gut are responsible for OSA-induced hypertension. OSA significantly increased systolic blood pressure at 7 and 14 days ( P<0.05), an effect that was abolished by the probiotic Clostridium butyricum or the prebiotic Hylon VII. The 16S rRNA analysis identified several short chain fatty acid-producing bacteria that were significantly increased by Cbutyricum and Hylon treatment. Acetate concentration in the cecum was decreased by 48% after OSA ( P<0.05), an effect that was prevented by Cbutyricum and Hylon. Cbutyricum and Hylon reduced OSA-induced dysbiosis, epithelial goblet cell loss, mucus barrier thinning, and activation of brain microglia ( P<0.05 for each). To examine the role of acetate in OSA-induced hypertension, we chronically infused acetate into the cecum during 2 weeks of sham or OSA. Restoring cecal acetate concentration prevented OSA-induced gut inflammation and hypertension ( P<0.05). These studies identify acetate as a key player in OSA-induced hypertension. We demonstrate that various methods to increase cecal acetate concentrations are protective from the adverse effects of OSA on the microbiota, gut, brain, and blood pressure. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
8. Bacteroides ovatusATCC 8483 monotherapy is superior to traditional fecal transplant and multi-strain bacteriotherapy in a murine colitis model
- Author
-
Ihekweazu, Faith D., Fofanova, Tatiana Y., Queliza, Karen, Nagy-Szakal, Dorottya, Stewart, Christopher J., Engevik, Melinda A., Hulten, Kristina G., Tatevian, Nina, Graham, David Y., Versalovic, James, Petrosino, Joseph F., and Kellermayer, Richard
- Abstract
ABSTRACTBackground and aimsBacteriotherapy aimed at addressing dysbiosis may be therapeutic for Inflammatory Bowel Diseases (IBDs). We sought to determine if defined Bacteroides-based bacteriotherapy could be an effective and consistent alternative to fecal microbiota transplantation (FMT) in a murine model of IBD.MethodsWe induced experimental colitis in 8– 12-week-old C57BL/6 mice using 2–3% dextran sodium sulfate. Mice were simultaneously treated by oral gavage with a triple-Bacteroidescocktail, individual Bacteroidesstrains, FMT using stool from healthy donor mice, or their own stool as a control. Survival, weight loss and markers of inflammation (histology, serum amyloid A, cytokine production) were correlated to 16S rRNAgene profiling of fecal and mucosal microbiomes.ResultsTriple-Bacteroidescombination therapy was more protective against weight loss and mortality than traditional FMT therapy. B. ovatusATCC8483 was more effective than any individual strain, or a combination of strains, in preventing weight loss, decreasing histological damage, dampening inflammatory response, and stimulating epithelial recovery. Irrespective of the treatment group, overall Bacteroidesabundance associated with treatment success and decreased cytokine production while the presence of Akkermansiacorrelated with treatment failure. However, the therapeutic benefit associated with high Bacteroidesabundance was negated in the presence of Streptococcus.ConclusionsBacteroides ovatusmonotherapy was more consistent and effective than traditional FMT at ameliorating colitis and stimulating epithelial recovery in a murine model of IBD. Given the tolerability of Bacteroides ovatusATCC 8483 in an active, on-going human study, this therapy may be repurposed for the management of IBD in a clinically expedient timeline.
- Published
- 2019
- Full Text
- View/download PDF
9. Leveraging Human Microbiome Features to Diagnose and Stratify Children with Irritable Bowel Syndrome
- Author
-
Hollister, Emily B., Oezguen, Numan, Chumpitazi, Bruno P., Luna, Ruth Ann, Weidler, Erica M., Rubio-Gonzales, Michelle, Dahdouli, Mahmoud, Cope, Julia L., Mistretta, Toni-Ann, Raza, Sabeen, Metcalf, Ginger A., Muzny, Donna M., Gibbs, Richard A., Petrosino, Joseph F., Heitkemper, Margaret, Savidge, Tor C., Shulman, Robert J., and Versalovic, James
- Abstract
Accurate diagnosis and stratification of children with irritable bowel syndrome (IBS) remain challenging. Given the central role of recurrent abdominal pain in IBS, we evaluated the relationships of pediatric IBS and abdominal pain with intestinal microbes and fecal metabolites using a comprehensive clinical characterization and multiomics strategy. Using rigorous clinical phenotyping, we identified preadolescent children (aged 7 to 12 years) with Rome III IBS (n= 23) and healthy controls (n= 22) and characterized their fecal microbial communities using whole-genome shotgun metagenomics and global unbiased fecal metabolomic profiling. Correlation-based approaches and machine learning algorithms identified associations between microbes, metabolites, and abdominal pain. IBS cases differed from controls with respect to key bacterial taxa (eg, Flavonifractor plautiiand Lachnospiraceae bacterium 7_1_58FAA), metagenomic functions (eg, carbohydrate metabolism and amino acid metabolism), and higher-order metabolites (eg, secondary bile acids, sterols, and steroid-like compounds). Significant associations between abdominal pain frequency and severity and intestinal microbial features were identified. A random forest classifier built on metagenomic and metabolic markers successfully distinguished IBS cases from controls (area under the curve, 0.93). Leveraging multiple lines of evidence, intestinal microbes, genes/pathways, and metabolites were associated with IBS, and these features were capable of distinguishing children with IBS from healthy children. These multi-omics features, and their links to childhood IBS coupled with nutritional interventions, may lead to new microbiome-guided diagnostic and therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
10. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases
- Author
-
Lloyd-Price, Jason, Arze, Cesar, Ananthakrishnan, Ashwin N., Schirmer, Melanie, Avila-Pacheco, Julian, Poon, Tiffany W., Andrews, Elizabeth, Ajami, Nadim J., Bonham, Kevin S., Brislawn, Colin J., Casero, David, Courtney, Holly, Gonzalez, Antonio, Graeber, Thomas G., Hall, A. Brantley, Lake, Kathleen, Landers, Carol J., Mallick, Himel, Plichta, Damian R., Prasad, Mahadev, Rahnavard, Gholamali, Sauk, Jenny, Shungin, Dmitry, Vázquez-Baeza, Yoshiki, White, Richard A., Braun, Jonathan, Denson, Lee A., Jansson, Janet K., Knight, Rob, Kugathasan, Subra, McGovern, Dermot P. B., Petrosino, Joseph F., Stappenbeck, Thaddeus S., Winter, Harland S., Clish, Clary B., Franzosa, Eric A., Vlamakis, Hera, Xavier, Ramnik J., and Huttenhower, Curtis
- Abstract
Inflammatory bowel diseases, which include Crohn’s disease and ulcerative colitis, affect several million individuals worldwide. Crohn’s disease and ulcerative colitis are complex diseases that are heterogeneous at the clinical, immunological, molecular, genetic, and microbial levels. Individual contributing factors have been the focus of extensive research. As part of the Integrative Human Microbiome Project (HMP2 or iHMP), we followed 132 subjects for one year each to generate integrated longitudinal molecular profiles of host and microbial activity during disease (up to 24 time points each; in total 2,965 stool, biopsy, and blood specimens). Here we present the results, which provide a comprehensive view of functional dysbiosis in the gut microbiome during inflammatory bowel disease activity. We demonstrate a characteristic increase in facultative anaerobes at the expense of obligate anaerobes, as well as molecular disruptions in microbial transcription (for example, among clostridia), metabolite pools (acylcarnitines, bile acids, and short-chain fatty acids), and levels of antibodies in host serum. Periods of disease activity were also marked by increases in temporal variability, with characteristic taxonomic, functional, and biochemical shifts. Finally, integrative analysis identified microbial, biochemical, and host factors central to this dysregulation. The study’s infrastructure resources, results, and data, which are available through the Inflammatory Bowel Disease Multi’omics Database (http://ibdmdb.org), provide the most comprehensive description to date of host and microbial activities in inflammatory bowel diseases.
- Published
- 2019
- Full Text
- View/download PDF
11. Early nasal microbiota and acute respiratory infections during the first years of life
- Author
-
Toivonen, Laura, Hasegawa, Kohei, Waris, Matti, Ajami, Nadim J, Petrosino, Joseph F, Camargo Jr, Carlos A, and Peltola, Ville
- Abstract
BackgroundEmerging evidence shows that airway microbiota may modulate local immune responses, thereby contributing to the susceptibility and severity of acute respiratory infections (ARIs). However, there are little data on the longitudinal relationships between airway microbiota and susceptibility to ARIs in children.ObjectiveWe aimed to investigate the association of early nasal microbiota and the subsequent risk of ARIs during the first years of life.MethodsIn this prospective population-based birth-cohort study in Finland, we followed 839 healthy infants for ARIs from birth to age 24 months. Nasal microbiota was tested using 16S rRNA gene sequencing at age 2 months. We applied an unsupervised clustering approach to identify early nasal microbiota profiles, and examined the association of profiles with the rate of ARIs during age 2–24 months.ResultsWe identified five nasal microbiota profiles dominated by Moraxella, Streptococcus, Dolosigranulum, Staphylococcusand Corynebacteriaceae, respectively. Incidence rate of ARIs was highest in children with an early Moraxella-dominant profile and lowest in those with a Corynebacteriaceae-dominant profile (738 vs 552/100 children years; unadjusted incidence rate ratio (IRR), 1.34; 95% CI 1.16 to 1.54; p < 0.001). After adjusting for nine potential confounders, the Moraxella-dominant profile-ARI association persisted (adjusted IRR (aIRR), 1.19; 95% CI 1.04 to 1.37; p = 0.01). Similarly, the incidence rate of lower respiratory tract infections (a subset of all ARIs) was significantly higher in children with an early Moraxella-dominant profile (aIRR, 2.79; 95% CI 1.04 to 8.09; p = 0.04).ConclusionMoraxella-dominant nasal microbiota profile in early infancy was associated with an increased rate of ARIs during the first 2 years of life.
- Published
- 2019
- Full Text
- View/download PDF
12. Temporal development of the gut microbiome in early childhood from the TEDDY study
- Author
-
Stewart, Christopher J., Ajami, Nadim J., O’Brien, Jacqueline L., Hutchinson, Diane S., Smith, Daniel P., Wong, Matthew C., Ross, Matthew C., Lloyd, Richard E., Doddapaneni, HarshaVardhan, Metcalf, Ginger A., Muzny, Donna, Gibbs, Richard A., Vatanen, Tommi, Huttenhower, Curtis, Xavier, Ramnik J., Rewers, Marian, Hagopian, William, Toppari, Jorma, Ziegler, Anette-G., She, Jin-Xiong, Akolkar, Beena, Lernmark, Ake, Hyoty, Heikki, Vehik, Kendra, Krischer, Jeffrey P., and Petrosino, Joseph F.
- Abstract
The development of the microbiome from infancy to childhood is dependent on a range of factors, with microbial–immune crosstalk during this time thought to be involved in the pathobiology of later life diseases1–9such as persistent islet autoimmunity and type 1 diabetes10–12. However, to our knowledge, no studies have performed extensive characterization of the microbiome in early life in a large, multi-centre population. Here we analyse longitudinal stool samples from 903 children between 3 and 46 months of age by 16S rRNA gene sequencing (n= 12,005) and metagenomic sequencing (n= 10,867), as part of the The Environmental Determinants of Diabetes in the Young (TEDDY) study. We show that the developing gut microbiome undergoes three distinct phases of microbiome progression: a developmental phase (months 3–14), a transitional phase (months 15–30), and a stable phase (months 31–46). Receipt of breast milk, either exclusive or partial, was the most significant factor associated with the microbiome structure. Breastfeeding was associated with higher levels of Bifidobacteriumspecies (B. breveand B. bifidum), and the cessation of breast milk resulted in faster maturation of the gut microbiome, as marked by the phylum Firmicutes. Birth mode was also significantly associated with the microbiome during the developmental phase, driven by higher levels of Bacteroidesspecies (particularly B. fragilis) in infants delivered vaginally. Bacteroideswas also associated with increased gut diversity and faster maturation, regardless of the birth mode. Environmental factors including geographical location and household exposures (such as siblings and furry pets) also represented important covariates. A nested case–control analysis revealed subtle associations between microbial taxonomy and the development of islet autoimmunity or type 1 diabetes. These data determine the structural and functional assembly of the microbiome in early life and provide a foundation for targeted mechanistic investigation into the consequences of microbial–immune crosstalk for long-term health.
- Published
- 2018
- Full Text
- View/download PDF
13. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study
- Author
-
Vatanen, Tommi, Franzosa, Eric A., Schwager, Randall, Tripathi, Surya, Arthur, Timothy D., Vehik, Kendra, Lernmark, Åke, Hagopian, William A., Rewers, Marian J., She, Jin-Xiong, Toppari, Jorma, Ziegler, Anette-G., Akolkar, Beena, Krischer, Jeffrey P., Stewart, Christopher J., Ajami, Nadim J., Petrosino, Joseph F., Gevers, Dirk, Lähdesmäki, Harri, Vlamakis, Hera, Huttenhower, Curtis, and Xavier, Ramnik J.
- Abstract
Type 1 diabetes (T1D) is an autoimmune disease that targets pancreatic islet beta cells and incorporates genetic and environmental factors1, including complex genetic elements2, patient exposures3and the gut microbiome4. Viral infections5and broader gut dysbioses6have been identified as potential causes or contributing factors; however, human studies have not yet identified microbial compositional or functional triggers that are predictive of islet autoimmunity or T1D. Here we analyse 10,913 metagenomes in stool samples from 783 mostly white, non-Hispanic children. The samples were collected monthly from three months of age until the clinical end point (islet autoimmunity or T1D) in the The Environmental Determinants of Diabetes in the Young (TEDDY) study, to characterize the natural history of the early gut microbiome in connection to islet autoimmunity, T1D diagnosis, and other common early life events such as antibiotic treatments and probiotics. The microbiomes of control children contained more genes that were related to fermentation and the biosynthesis of short-chain fatty acids, but these were not consistently associated with particular taxa across geographically diverse clinical centres, suggesting that microbial factors associated with T1D are taxonomically diffuse but functionally more coherent. When we investigated the broader establishment and development of the infant microbiome, both taxonomic and functional profiles were dynamic and highly individualized, and dominated in the first year of life by one of three largely exclusive Bifidobacteriumspecies (B. bifidum, B. breveor B. longum) or by the phylum Proteobacteria. In particular, the strain-specific carriage of genes for the utilization of human milk oligosaccharide within a subset of B. longumwas present specifically in breast-fed infants. These analyses of TEDDY gut metagenomes provide, to our knowledge, the largest and most detailed longitudinal functional profile of the developing gut microbiome in relation to islet autoimmunity, T1D and other early childhood events. Together with existing evidence from human cohorts7,8and a T1D mouse model9, these data support the protective effects of short-chain fatty acids in early-onset human T1D.
- Published
- 2018
- Full Text
- View/download PDF
14. Role of the Gut Microbiome in Obstructive Sleep Apnea-Induced Hypertension.
- Author
-
Durgan, David J., Ganesh, Bhanu P., Cope, Julia L., Ajami, Nadim J., Phillips, Sharon C., Petrosino, Joseph F., Hollister, Emily B., Bryan Jr, Robert M., and Bryan, Robert M Jr
- Abstract
Individuals suffering from obstructive sleep apnea (OSA) are at increased risk for systemic hypertension. The importance of a healthy gut microbiota, and detriment of a dysbiotic microbiota, on host physiology is becoming increasingly evident. We tested the hypothesis that gut dysbiosis contributes to hypertension observed with OSA. OSA was modeled in rats by inflating a tracheal balloon during the sleep cycle (10-s inflations, 60 per hour). On normal chow diet, OSA had no effect on blood pressure; however, in rats fed a high-fat diet, blood pressure increased 24 and 29 mm Hg after 7 and 14 days of OSA, respectively (P<0.05 each). Bacterial community characterization was performed on fecal pellets isolated before and after 14 days of OSA in chow and high-fat fed rats. High-fat diet and OSA led to significant alterations of the gut microbiota, including decreases in bacterial taxa known to produce the short chain fatty acid butyrate (P<0.05). Finally, transplant of dysbiotic cecal contents from hypertensive OSA rats on high-fat diet into OSA recipient rats on normal chow diet (shown to be normotensive) resulted in hypertension similar to that of the donor (increased 14 and 32 mm Hg after 7 and 14 days of OSA, respectively; P<0.05). These studies demonstrate a causal relationship between gut dysbiosis and hypertension, and suggest that manipulation of the microbiota may be a viable treatment for OSA-induced, and possibly other forms of, hypertension. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
15. Association of Maternal Gestational Weight Gain With the Infant Fecal Microbiota
- Author
-
Robinson, Alyssa, Fiechtner, Lauren, Roche, Brianna, Ajami, Nadim J., Petrosino, Joseph F., Camargo, Carlos A., Taveras, Elsie M., and Hasegawa, Kohei
- Abstract
Pregnancy characteristics may influence the infant fecal microbiota during early life. We aimed to examine associations of maternal gestational weight gain with infant fecal microbiota composition, bacterial community richness, and Shannon diversity index. We analyzed data from a prospective cohort study of healthy infants. We collected prenatal data, including report of mother's gestational weight gain, and infant fecal samples from 84 infant-mother dyads. By applying 16S rRNA gene sequencing and an unbiased clustering by partitioning around medoids using Bray-Curtis distances, we identified 4 fecal microbiota profiles, and examined the associations of maternal gestational weight gain with the 4 fecal microbiota profiles, bacterial community richness, and Shannon diversity index. Overall, the median age of infants was 4.0 months and 43% were girls. The mothers of the 84 infants gained a mean of 14.2 kg (standard deviation, 5.4 kg) during pregnancy. We identified 4 distinct microbiota profiles: Bifidobacterium-dominant (42%), Enterobacter/Veillonella-dominant (23%), Bacteroides-dominant (19%), and Escherichia-dominant (17%). Infants whose mothers had higher gestational weight gain were less likely to have a Bacteroides-dominant profile, corresponding to a relative risk ratio of 0.83 (95% confidence interval, 0.71–0.96; P= 0.01) per 1 kg increase in weight. In addition, higher gestational weight gain was also associated with lower bacterial community richness and Shannon diversity index (P< 0.05). In this prospective cohort study of healthy infants, maternal gestational weight gain was associated with the infant fecal microbiota profiles, bacterial community richness, and Shannon diversity index.
- Published
- 2017
- Full Text
- View/download PDF
16. Association of Maternal Gestational Weight Gain With the Infant Fecal Microbiota
- Author
-
Robinson, Alyssa, Fiechtner, Lauren, Roche, Brianna, Ajami, Nadim J., Petrosino, Joseph F., Camargo, Carlos A., Taveras, Elsie M., and Hasegawa, Kohei
- Abstract
Supplemental Digital Content is available in the text
- Published
- 2017
- Full Text
- View/download PDF
17. Nasal Airway Microbiota Profile and Severe Bronchiolitis in Infants
- Author
-
Hasegawa, Kohei, Linnemann, Rachel W., Mansbach, Jonathan M., Ajami, Nadim J., Espinola, Janice A., Petrosino, Joseph F., Piedra, Pedro A., Stevenson, Michelle D., Sullivan, Ashley F., Thompson, Amy D., and Camargo, Carlos A.
- Published
- 2017
- Full Text
- View/download PDF
18. Successful collection of stool samples for microbiome analyses from a large community-based population of elderly men
- Author
-
Abrahamson, Melanie, Hooker, Elizabeth, Ajami, Nadim J., Petrosino, Joseph F., and Orwoll, Eric S.
- Abstract
The relationship of the gastrointestinal microbiome to health and disease is of major research interest, including the effects of the gut microbiota on age related conditions. Here we report on the outcome of a project to collect stool samples on a large number of community dwelling elderly men using the OMNIgene-GUT stool/feces collection kit (OMR-200, DNA Genotek, Ottawa, Canada). Among 1328 men who were eligible for stool collection, 982 (74%) agreed to participate and 951 submitted samples. The collection process was reported to be acceptable, almost all samples obtained were adequate, the process of sample handling by mail was uniformly successful. The DNA obtained provided excellent results in microbiome analyses, yielding an abundance of species and a diversity of taxa as would be predicted. Our results suggest that population studies of older participants involving remote stool sample collection are feasible. These approaches would allow large scale research projects of the association of the gut microbiota with important clinical outcomes.
- Published
- 2017
- Full Text
- View/download PDF
19. Gut microbiota as a source of a surrogate antigen that triggers autoimmunity in an immune privileged site
- Author
-
Zárate-Bladés, Carlos R., Horai, Reiko, Mattapallil, Mary J., Ajami, Nadim J., Wong, Matthew, Petrosino, Joseph F., Itoh, Kikuji, Chan, Chi-Chao, and Caspi, Rachel R.
- Published
- 2017
- Full Text
- View/download PDF
20. Maternal gut microbiota mediate intergenerational effects of high-fat diet on descendant social behavior
- Author
-
Di Gesù, Claudia M., Matz, Lisa M., Bolding, Ian J., Fultz, Robert, Hoffman, Kristi L., Gammazza, Antonella Marino, Petrosino, Joseph F., and Buffington, Shelly A.
- Published
- 2023
- Full Text
- View/download PDF
21. Decreased microbiota diversity associated with urinary tract infection in a trial of bacterial interference.
- Author
-
Horwitz, Deborah, McCue, Tyler, Mapes, Abigail C., Ajami, Nadim J., Petrosino, Joseph F., Ramig, Robert F., and Trautner, Barbara W.
- Abstract
Summary Background Patients with long-term indwelling catheters are at high risk of catheter-associated urinary tract infection (CAUTI). We hypothesized that colonizing the bladder with a benign Escherichia coli strain ( E. coli HU2117, a derivative of E. coli 83972) would prevent CAUTI in older, catheterized adults. Materials and methods Adults with chronic, indwelling urinary catheters received study catheters that had been pre-coated with E. coli HU2117. We monitored the cultivatable organisms in the bladder for 28 days or until loss of E. coli HU2117. Urine from 4 subjects was collected longitudinally for 16S rRNA gene profiling. Results Eight of the ten subjects (average age 70.9 years) became colonized with E. coli HU2117, with a mean duration of 57.7 days (median: 28.5, range 0–266). All subjects also remained colonized by uropathogens. Five subjects suffered invasive UTI, 3 febrile UTI and 2 urosepsis/bacteremia, all associated with overgrowth of a urinary pathogen. Colonization with E. coli HU2117 did not impact bacterial bladder diversity, but subjects who developed infections had less diverse bladder microbiota. Conclusions Colonization with E. coli HU2117 did not prevent bladder colonization or subsequent invasive disease by uropathogens. Microbial diversity may play a protective role against invasive infection of the catheterized bladder. Trial Registration: ClinicalTrials.gov , NCT00554996 http://clinicaltrials.gov/ct2/show/NCT00554996 . [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
22. Microbiome–Epigenome Interactions and the Environmental Origins of Inflammatory Bowel Diseases
- Author
-
Fofanova, Tatiana Y., Petrosino, Joseph F., and Kellermayer, Richard
- Published
- 2016
- Full Text
- View/download PDF
23. Microbiome–Epigenome Interactions and the Environmental Origins of Inflammatory Bowel Diseases
- Author
-
Fofanova, Tatiana Y., Petrosino, Joseph F., and Kellermayer, Richard
- Abstract
The incidence of pediatric inflammatory bowel disease (IBD), which includes Crohn disease and ulcerative colitis, has risen alarmingly in the Western and developing world in recent decades. Epidemiologic (including monozygotic twin and migrant) studies highlight the substantial role of environment and nutrition in IBD etiology. Here we review the literature supporting the developmental and environmental origins hypothesis of IBD. We also provide a detailed exploration of how the human microbiome and epigenome (primarily through DNA methylation) may be important elements in the developmental origins of IBD in both children and adults.
- Published
- 2016
- Full Text
- View/download PDF
24. Development and Accuracy of Quantitative Real-Time Polymerase Chain Reaction Assays for Detection and Quantification of Enterotoxigenic Escherichia coli (ETEC) Heat Labile and Heat Stable Toxin Genes in Travelers' Diarrhea Samples.
- Author
-
Youmans, Bonnie P., Ajami, Nadim J., Zhi-Dong Jiang, Petrosino, Joseph F., DuPont, Herbert L., and Highlander, Sarah K.
- Published
- 2014
- Full Text
- View/download PDF
25. Maternal gut microbiota mediate intergenerational effects of high-fat diet on descendant social behavior
- Author
-
Di Gesù, Claudia M., Matz, Lisa M., Bolding, Ian J., Fultz, Robert, Hoffman, Kristi L., Gammazza, Antonella Marino, Petrosino, Joseph F., and Buffington, Shelly A.
- Abstract
Dysbiosis of the maternal gut microbiome during pregnancy is associated with adverse neurodevelopmental outcomes. We previously showed that maternal high-fat diet (MHFD) in mice induces gut dysbiosis, social dysfunction, and underlying synaptic plasticity deficits in male offspring (F1). Here, we reason that, if HFD-mediated changes in maternal gut microbiota drive offspring social deficits, then MHFD-induced dysbiosis in F1female MHFD offspring would likewise impair F2social behavior. Metataxonomic sequencing reveals reduced microbial richness among female F1MHFD offspring. Despite recovery of microbial richness among MHFD-descendant F2mice, they display social dysfunction. Post-weaning Limosilactobacillus reuteritreatment increases the abundance of short-chain fatty acid-producing taxa and rescues MHFD-descendant F2social deficits. L. reuteriexerts a sexually dimorphic impact on gut microbiota configuration, increasing discriminant taxa between female cohorts. Collectively, these results show multigenerational impacts of HFD-induced dysbiosis in the maternal lineage and highlight the potential of maternal microbiome-targeted interventions for neurodevelopmental disorders.
- Published
- 2022
- Full Text
- View/download PDF
26. Murine Model of Chemotherapy-Induced Extraintestinal Pathogenic Escherichia coliTranslocation
- Author
-
Green, Sabrina I., Ajami, Nadim J., Ma, Li, Poole, Nina M., Price, Roger E., Petrosino, Joseph F., and Maresso, Anthony W.
- Abstract
ABSTRACTEscherichia coliis a major cause of life-threatening infections in patients with neutropenia, particularly those receiving chemotherapy for the treatment of cancer. In most cases, these infections originate from opportunistic strains living within the patient's gastrointestinal tract which then translocate to major organ systems. There are no animal models that faithfully recapitulate these infections, and, as such, the host or bacterial factors that govern this process remain unidentified. We present here a novel model of chemotherapy-induced bacterial translocation of E. coli. Oral gavage of BALB/c mice with a clinical isolate of extraintestinal pathogenic E. coli(ExPEC) leads to stable and long-term colonization of the murine intestine. Following the induction of neutropenia with the chemotherapeutic drug cyclophosphamide, ExPEC translocates from the intestine to the lungs, liver, spleen, and kidneys with concomitant morbidity in infected animals. Translocation can also occur in mice bearing mammary tumors, even in the absence of chemotherapy. Translocation of ExPEC is also associated with an increase of the diversity of bacterial DNA detected in the blood. This is the first report of a chemotherapy-based animal model of ExPEC translocation in cancerous mice, a system that can be readily used to identify important virulence factors for this process.
- Published
- 2015
- Full Text
- View/download PDF
27. Characterization of the human gut microbiome during travelers' diarrhea
- Author
-
Youmans, Bonnie P, Ajami, Nadim J, Jiang, Zhi-Dong, Campbell, Frederick, Wadsworth, W Duncan, Petrosino, Joseph F, DuPont, Herbert L, and Highlander, Sarah K
- Abstract
Alterations in the gut microbiota are correlated with ailments such as obesity, inflammatory bowel disease, and diarrhea. Up to 60% of individuals traveling from industrialized to developing countries acquire a form of secretory diarrhea known as travelers' diarrhea (TD), and enterotoxigenic Escherichia coli(ETEC) and norovirus (NoV) are the leading causative pathogens. Presumably, TD alters the gut microbiome, however the effect of TD on gut communities has not been studied. We report the first analysis of bacterial gut populations associated with TD. We examined and compared the gut microbiomes of individuals who developed TD associated with ETEC, NoV, or mixed pathogens, and TD with no pathogen identified, to healthy travelers. We observed a signature dysbiotic gut microbiome profile of high Firmicutes:Bacteroidetes ratios in the travelers who developed diarrhea, regardless of etiologic agent or presence of a pathogen. There was no significant difference in α-diversity among travelers. The bacterial composition of the microbiota of the healthy travelers was similar to the diarrheal groups, however the β-diversity of the healthy travelers was significantly different than any pathogen-associated TD group. Further comparison of the healthy traveler microbiota to those from healthy subjects who were part of the Human Microbiome Project also revealed a significantly higher Firmicutes:Bacteriodetes ratio in the healthy travelers and significantly different β-diversity. Thus, the composition of the gut microbiome in healthy, diarrhea-free travelers has characteristics of a dysbiotic gut, suggesting that these alterations could be associated with factors such as travel.
- Published
- 2015
- Full Text
- View/download PDF
28. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis.
- Author
-
Noval Rivas, Magali, Burton, Oliver T., Wise, Petra, Zhang, Yu-qian, Hobson, Suejy A., Garcia Lloret, Maria, Chehoud, Christel, Kuczynski, Justin, DeSantis, Todd, Warrington, Janet, Hyde, Embriette R., Petrosino, Joseph F., Gerber, Georg K., Bry, Lynn, Oettgen, Hans C., Mazmanian, Sarkis K., and Chatila, Talal A.
- Subjects
FOOD allergy ,ANAPHYLAXIS ,IMMUNOLOGICAL tolerance ,MICROBIAL ecology ,T cells ,GENETIC mutation ,INFECTIOUS disease transmission ,DISEASE susceptibility - Abstract
Background: Commensal microbiota play a critical role in maintaining oral tolerance. The effect of food allergy on the gut microbial ecology remains unknown. Objective: We sought to establish the composition of the gut microbiota in experimental food allergy and its role in disease pathogenesis. Methods: Food allergy–prone mice with a gain-of-function mutation in the IL-4 receptor α chain (Il4raF709) and wild-type (WT) control animals were subjected to oral sensitization with chicken egg ovalbumin (OVA). Enforced tolerance was achieved by using allergen-specific regulatory T (Treg) cells. Community structure analysis of gut microbiota was performed by using a high-density 16S rDNA oligonucleotide microarrays (PhyloChip) and massively parallel pyrosequencing of 16S rDNA amplicons. Results: OVA-sensitized Il4raF709 mice exhibited a specific microbiota signature characterized by coordinate changes in the abundance of taxa of several bacterial families, including the Lachnospiraceae, Lactobacillaceae, Rikenellaceae, and Porphyromonadaceae. This signature was not shared by similarly sensitized WT mice, which did not exhibit an OVA-induced allergic response. Treatment of OVA-sensitized Il4raF709 mice with OVA-specific Treg cells led to a distinct tolerance-associated signature coincident with the suppression of the allergic response. The microbiota of allergen-sensitized Il4raF709 mice differentially promoted OVA-specific IgE responses and anaphylaxis when reconstituted in WT germ-free mice. Conclusion: Mice with food allergy exhibit a specific gut microbiota signature capable of transmitting disease susceptibility and subject to reprogramming by enforced tolerance. Disease-associated microbiota may thus play a pathogenic role in food allergy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
29. Gastrointestinal Microbiome Signatures of Pediatric Patients With Irritable Bowel Syndrome.
- Author
-
Saulnier, Delphine M., Riehle, Kevin, Mistretta, Toni–Ann, Diaz, Maria–Alejandra, Mandal, Debasmita, Raza, Sabeen, Weidler, Erica M., Qin, Xiang, Coarfa, Cristian, Milosavljevic, Aleksandar, Petrosino, Joseph F., Highlander, Sarah, Gibbs, Richard, Lynch, Susan V., Shulman, Robert J., and Versalovic, James
- Subjects
GUT microbiome ,IRRITABLE colon ,JUVENILE diseases ,RNA ,HAEMOPHILUS parainfluenzae ,GASTROINTESTINAL diseases ,ABDOMINAL pain in children ,DISEASE relapse - Abstract
Background & Aims: The intestinal microbiomes of healthy children and pediatric patients with irritable bowel syndrome (IBS) are not well defined. Studies in adults have indicated that the gastrointestinal microbiota could be involved in IBS. Methods: We analyzed 71 samples from 22 children with IBS (pediatric Rome III criteria) and 22 healthy children, ages 7–12 years, by 16S ribosomal RNA gene sequencing, with an average of 54,287 reads/stool sample (average 454 read length = 503 bases). Data were analyzed using phylogenetic-based clustering (Unifrac), or an operational taxonomic unit (OTU) approach using a supervised machine learning tool (randomForest). Most samples were also hybridized to a microarray that can detect 8741 bacterial taxa (16S rRNA PhyloChip). Results: Microbiomes associated with pediatric IBS were characterized by a significantly greater percentage of the class γ-proteobacteria (0.07% vs 0.89% of total bacteria, respectively; P < .05); 1 prominent component of this group was Haemophilus parainfluenzae. Differences highlighted by 454 sequencing were confirmed by high-resolution PhyloChip analysis. Using supervised learning techniques, we were able to classify different subtypes of IBS with a success rate of 98.5%, using limited sets of discriminant bacterial species. A novel Ruminococcus-like microbe was associated with IBS, indicating the potential utility of microbe discovery for gastrointestinal disorders. A greater frequency of pain correlated with an increased abundance of several bacterial taxa from the genus Alistipes. Conclusions: Using16S metagenomics by PhyloChip DNA hybridization and deep 454 pyrosequencing, we associated specific microbiome signatures with pediatric IBS. These findings indicate the important association between gastrointestinal microbes and IBS in children; these approaches might be used in diagnosis of functional bowel disorders in pediatric patients. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
30. The Genome of Th17 Cell-Inducing Segmented Filamentous Bacteria Reveals Extensive Auxotrophy and Adaptations to the Intestinal Environment.
- Author
-
Sczesnak, Andrew, Segata, Nicola, Qin, Xiang, Gevers, Dirk, Petrosino, Joseph F., Huttenhower, Curtis, Littman, Dan R., and Ivanov, Ivaylo I.
- Subjects
GUT microbiome ,T cells ,NUCLEOTIDE sequence ,COMPARATIVE studies ,HOST-bacteria relationships ,BACTERIAL genetics ,COMPLEMENT (Immunology) - Abstract
Summary: Perturbations of the composition of the symbiotic intestinal microbiota can have profound consequences for host metabolism and immunity. In mice, segmented filamentous bacteria (SFB) direct the accumulation of potentially proinflammatory Th17 cells in the intestinal lamina propria. We present the genome sequence of SFB isolated from monocolonized mice, which classifies SFB phylogenetically as a unique member of Clostridiales with a highly reduced genome. Annotation analysis demonstrates that SFB depend on their environment for amino acids and essential nutrients and may utilize host and dietary glycans for carbon, nitrogen, and energy. Comparative analyses reveal that SFB are functionally related to members of the genus Clostridium and several pathogenic or commensal “minimal” genera, including Finegoldia, Mycoplasma, Borrelia, and Phytoplasma. However, SFB are functionally distinct from all 1200 examined genomes, indicating a gene complement representing biology relatively unique to their role as a gut commensal closely tied to host metabolism and immunity. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
31. Monotonous Diets Protect Against Acute Colitis in Mice
- Author
-
Nagy-Szakal, Dorottya, Mir, Sabina A.V., Ross, Matthew C., Tatevian, Nina, Petrosino, Joseph F., and Kellermayer, Richard
- Abstract
Multiple characteristics of industrialization have been proposed to contribute to the global emergence of inflammatory bowel diseases (IBDs: Crohn disease and ulcerative colitis). Major changes in eating habits during the last decades and the effectiveness of exclusive enteral nutrition in the treatment of Crohn disease indicate the etiologic importance of dietary intake in IBDs. A uniform characteristic of nutrition in developing countries (where the incidence of IBD is low) and exclusive enteral nutrition is their consistent nature for prolonged periods; however, the potentially beneficial effect of dietary monotony in respect to mammalian intestinal inflammation has not been examined.
- Published
- 2013
- Full Text
- View/download PDF
32. Monotonous Diets Protect Against Acute Colitis in Mice
- Author
-
Nagy-Szakal, Dorottya, Mir, Sabina A.V., Ross, Matthew C., Tatevian, Nina, Petrosino, Joseph F., and Kellermayer, Richard
- Abstract
Multiple characteristics of industrialization have been proposed to contribute to the global emergence of inflammatory bowel diseases (IBDs: Crohn disease and ulcerative colitis). Major changes in eating habits during the last decades and the effectiveness of exclusive enteral nutrition in the treatment of Crohn disease indicate the etiologic importance of dietary intake in IBDs. A uniform characteristic of nutrition in developing countries (where the incidence of IBD is low) and exclusive enteral nutrition is their consistent nature for prolonged periods; however, the potentially beneficial effect of dietary monotony in respect to mammalian intestinal inflammation has not been examined. The association between alternating (2 different complete chows) and persistent regular diets, and dextran sulfate sodium colitis susceptibility in C57BL/6J mice was studied. Colonic mucosal microbiota changes were investigated by high-throughput pyrosequencing of the 16S rRNAgene. The severity of colitis increased upon dietary alternation compared with consistent control feeding. The microbiota of the alternating nutritional group clustered discretely from both control groups. Our findings highlight that monotonous dietary intake may decrease mammalian vulnerability against colitis in association with microbiota separation. The epidemiologic and therapeutic implications of our results are also discussed.
- Published
- 2013
- Full Text
- View/download PDF
33. Maternal micronutrients can modify colonic mucosal microbiota maturation in murine offspring
- Author
-
Nagy-Szakal, Dorottya, Ross, Matthew C., Dowd, Scot E., Mir, Sabina A.V., Schaible, Tiffany D., Petrosino, Joseph F., and Kellermayer, Richard
- Abstract
Epidemiologic data suggest that early nutritional exposures may inflict persistent changes in the developing mammalian “super-organism” (i.e., the host and its residing microbiota). Such persistent modifications could predispose young adults to inflammatory bowel diseases (IBD). We recently observed that the dietary supplementation of four micronutrients to dams augmented colitis susceptibility in murine offspring in association with mucosal microbiota composition changes. In this study the effects of the four micronutrients on the microbiota of dams and female mice was examined. Additionally, age dependent microbiota composition shifts during pediatric development were delineated from the previous offspring data sets. Maternal and adult female microbiota did not separate secondary to the nutritional intervention. Significant microbiota composition changes occurred from postnatal day 30 (P30) to P90 at the level of 1 phylum and 15 genera. Most of these changes were absent or opposite in the maternally supplemented offspring. Nutritionally induced alterations in mucosal microbiota maturation may be contributors to colitis susceptibility in mammals.
- Published
- 2012
- Full Text
- View/download PDF
34. Host Response to Probiotics Determined by Nutritional Status of Rotavirus-infected Neonatal Mice
- Author
-
Preidis, Geoffrey A., Saulnier, Delphine M., Blutt, Sarah E., Mistretta, Toni-Ann, Riehle, Kevin P., Major, Angela M., Venable, Susan F., Barrish, James P., Finegold, Milton J., Petrosino, Joseph F., Guerrant, Richard L., Conner, Margaret E., and Versalovic, James
- Abstract
Beneficial microbes and probiotics are promising agents for the prevention and treatment of enteric and diarrheal diseases in children; however, little is known about their in vivo mechanisms of action. We used a neonatal mouse model of rotavirus diarrhea to gain insight into how probiotics ameliorate acute gastroenteritis.
- Published
- 2012
- Full Text
- View/download PDF
35. Host Response to Probiotics Determined by Nutritional Status of Rotavirus-infected Neonatal Mice
- Author
-
Preidis, Geoffrey A., Saulnier, Delphine M., Blutt, Sarah E., Mistretta, Toni-Ann, Riehle, Kevin P., Major, Angela M., Venable, Susan F., Barrish, James P., Finegold, Milton J., Petrosino, Joseph F., Guerrant, Richard L., Conner, Margaret E., and Versalovic, James
- Abstract
Beneficial microbes and probiotics are promising agents for the prevention and treatment of enteric and diarrheal diseases in children; however, little is known about their in vivo mechanisms of action. We used a neonatal mouse model of rotavirus diarrhea to gain insight into how probiotics ameliorate acute gastroenteritis. Rotavirus-infected mice were treated with 1 of 2 strains of human-derived Lactobacillus reuteri. We assessed intestinal microbiome composition with 16S metagenomic sequencing, enterocyte migration and proliferation with 5-bromo-2'-deoxyuridine, and antibody and cytokine concentrations with multiplex analyses of intestinal explant cultures. Probiotics reduced diarrhea duration, improved intestinal histopathology, and enhanced intestinal microbiome richness and phylogenetic diversity. The magnitude of reduction of diarrhea by probiotics was strain specific and influenced by nutritional status. L reuteriDSM 17938 reduced diarrhea duration by 0, 1, and 2 days in underweight, normal weight, and overweight pups, respectively. The magnitude of reduction of diarrhea duration correlated with increased enterocyte proliferation and migration. Strain ATCC PTA 6475 reduced diarrhea duration by 1 day in all of the mice without increasing enterocyte proliferation. Both probiotic strains decreased concentrations of proinflammatory cytokines, including macrophage inflammatory protein-1a and interleukin-1ß, in all of the animals, and increased rotavirus-specific antibodies in all but the underweight animals. Body weight also influenced the host response to rotavirus, in terms of diarrhea duration, enterocyte turnover, and antibody production. These data suggest that probiotic enhancement of enterocyte proliferation, villus repopulation, and virus-specific antibodies may contribute to diarrhea resolution, and that nutritional status influences the host response to both beneficial microbes and pathogens.
- Published
- 2012
- Full Text
- View/download PDF
36. Stabilization of the murine gut microbiome following weaning
- Author
-
Schloss, Patrick D., Schubert, Alyxandria M., Zackular, Joseph P., Iverson, Kathryn D., Young, Vincent B., and Petrosino, Joseph F.
- Abstract
Ecologists hypothesize that community structure and stability affect productivity, sensitivity to invasion and extinction, and resilience and resistance to perturbations. Viewed in the context of the gut microbiome, the stability of the gut community is important for understanding the effects of antibiotics, diet change and other perturbations on host health and colonization resistance. Here we describe the dynamics of a self-contained community, the murine gut microbiome. Using 16S rRNA gene sequencing of fecal samples collected daily from individual mice, we characterized the community membership and structure to determine whether there were significant changes in the gut community during the first year of life. Based on analysis of molecular variance, we observed two community states. The first was observed in the 10 days following weaning and the second was observed by 15 days following weaning. Interestingly, these two states had the same bacterial populations, but those populations had different relative abundances in the two states. By calculating the root mean squared distances between samples collected in the early and late states for each mouse, we observed that the late state was more stable than the early state. This increase in stability was not correlated with increased taxonomic richness, taxonomic diversity, or phylogenetic diversity. In the absence of an experimentally induced perturbation, the second community state was relatively constant through 364 days post weaning. These results suggest a high degree of stability in the microbiome once the community reached the second state.
- Published
- 2012
- Full Text
- View/download PDF
37. Opinion Paper: Promise and Pragmatism in Clinical Microbiome Research
- Author
-
Ajami, Nadim J., Hutchinson, Diane S., and Petrosino, Joseph F.
- Abstract
The evolution of human microbiome research has lead to a systems biology approach that encompasses multidisciplinary investigations. The implementation of next generation sequencing technologies has allowed researchers to study unculturable organisms, discover novel ones, and provide insights into the role of the human microbiome in health and disease. When these approaches are applied to large-scale longitudinal studies designed to interrogate the association of the microbiome with specific clinical outcomes, the development of new therapeutics and diagnostics intended to modulate or detect changes in microbiome composition to improve human health are born. We are just starting to unravel the role of the microbiome in a wide-variety of diseases, and while some of it appears to be related to causation and provide opportunities for intervention, a good dose of pragmatism is warranted as the field is still in its infancy.
- Published
- 2016
38. Chromosomal System for Studying AmpC-Mediated β-Lactam Resistance Mutation in Escherichia coli
- Author
-
Petrosino, Joseph F., Pendleton, Amanda R., Weiner, Joel H., and Rosenberg, Susan M.
- Abstract
ABSTRACTIn some enterobacterial pathogens, but not in Escherichia coli, loss-of-function mutations in the ampDgene are a common route to β-lactam antibiotic resistance. We constructed an assay system for studying mechanism(s) of enterobacterial ampDmutation using the well-developed genetics of E. coli. We integrated the Enterobacter ampRCgenes into the E. colichromosome. These cells acquire spontaneous recombination- and SOS response-independent β-lactam resistance mutations in ampD. This chromosomal system is useful for studying mutation mechanisms that promote antibiotic resistance.
- Published
- 2002
- Full Text
- View/download PDF
39. Susceptibility of β-lactamase to core amino acid substitutions
- Author
-
Petrosino, Joseph F., Baker, Matthew, and Palzkill, Timothy
- Abstract
To determine which amino acids in TEM-1 β-lactamase are important for its structure and function, random libraries were previously constructed which systematically randomized the 263 codons of the mature enzyme. A comprehensive screening of these libraries identified several TEM-1 β-lactamase core positions, including F66 and L76, which are strictly required for wild-type levels of hydrolytic activity. An examination of positions 66 and 76 in the class A β-lactamase gene family shows that a phenylalanine at position 66 is strongly conserved while position 76 varies considerably among other β-lactamases. It is possible that position 76 varies in the gene family because β-lactamase mutants with non-conservative substitutions at position 76 retain partial function. In contrast, position 66 may remain unchanged in the gene family because non-conservative substitutions at this location are detrimental for enzyme structure and function. By determining the β-lactam resistance levels of the 38 possible mutants at positions 66 and 76 in the TEM-1 enzyme, it was confirmed that position 76 is indeed more tolerant of non-conservative substitutions. An analysis of the Protein Data Bank files for three class A β-lactamases indicates that volume constraints at position 66 are at least partly responsible for the low tolerance of substitutions at this position.
- Published
- 1999
40. Increased Moraxella and Streptococcus species abundance after severe bronchiolitis is associated with recurrent wheezing.
- Author
-
Mansbach, Jonathan M., Luna, Pamela N., Shaw, Chad A., Hasegawa, Kohei, Petrosino, Joseph F., Piedra, Pedro A., Sullivan, Ashley F., Espinola, Janice A., Stewart, Christopher J., and Camargo, Carlos A.
- Abstract
The role of the airway microbiome in the development of recurrent wheezing and asthma remains uncertain, particularly in the high-risk group of infants hospitalized for bronchiolitis. We sought to examine the relation of the nasal microbiota at bronchiolitis-related hospitalization and 3 later points to the risk of recurrent wheezing by age 3 years. In 17 US centers researchers collected clinical data and nasal swabs from infants hospitalized for bronchiolitis. Trained parents collected nasal swabs 3 weeks after hospitalization and, when healthy, during the summer and 1 year after hospitalization. We applied 16S rRNA gene sequencing to all nasal swabs. We used joint modeling to examine the relation of longitudinal nasal microbiota abundances to the risk of recurrent wheezing. Among 842 infants hospitalized for bronchiolitis, there was 88% follow-up at 3 years, and 31% had recurrent wheezing. The median age at enrollment was 3.2 months (interquartile range, 1.7-5.8 months). In joint modeling analyses adjusting for 16 covariates, including viral cause, a 10% increase in relative abundance of Moraxella or Streptococcus species 3 weeks after day 1 of hospitalization was associated with an increased risk of recurrent wheezing (hazard ratio [HR] of 1.38 and 95% high-density interval [HDI] of 1.11-1.85 and HR of 1.76 and 95% HDI of 1.13-3.19, respectively). Increased Streptococcus species abundance the summer after hospitalization was also associated with a greater risk of recurrent wheezing (HR, 1.76; 95% HDI, 1.15-3.27). Enrichment of Moraxella or Streptococcus species after bronchiolitis hospitalization was associated with recurrent wheezing by age 3 years, possibly providing new avenues to ameliorate the long-term respiratory outcomes of infants with severe bronchiolitis. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
41. Impact of Diabetes on the Gut and Salivary IgA Microbiomes
- Author
-
Brown, Eric L., Essigmann, Heather T., Hoffman, Kristi L., Palm, Noah W., Gunter, Sarah M., Sederstrom, Joel M., Petrosino, Joseph F., Jun, Goo, Aguilar, David, Perkison, William B., Hanis, Craig L., and DuPont, Herbert L.
- Abstract
Mucosal surfaces like those present in the lung, gut, and mouth interface with distinct external environments. These mucosal gateways are not only portals of entry for potential pathogens but also homes to microbial communities that impact host health. Secretory immunoglobulin A (SIgA) is the single most abundant acquired immune component secreted onto mucosal surfaces and, via the process of immune exclusion, shapes the architecture of these microbiomes. Not all microorganisms at mucosal surfaces are targeted by SIgA; therefore, a better understanding of the SIgA-coated fraction may identify the microbial constituents that stimulate host immune responses in the context of health and disease.
- Published
- 2020
- Full Text
- View/download PDF
42. Serum cathelicidin, nasopharyngeal microbiota, and disease severity among infants hospitalized with bronchiolitis.
- Author
-
Hasegawa, Kohei, Mansbach, Jonathan M., Ajami, Nadim J., Petrosino, Joseph F., Freishtat, Robert J., Teach, Stephen J., Piedra, Pedro A., and Jr.Camargo, Carlos A.
- Published
- 2017
- Full Text
- View/download PDF
43. Respiratory syncytial virus and rhinovirus severe bronchiolitis are associated with distinct nasopharyngeal microbiota.
- Author
-
Mansbach, Jonathan M., Hasegawa, Kohei, Henke, David M., Ajami, Nadim J., Petrosino, Joseph F., Shaw, Chad A., Piedra, Pedro A., Sullivan, Ashley F., Espinola, Janice A., and Jr.Camargo, Carlos A.
- Published
- 2016
- Full Text
- View/download PDF
44. The Gut Microbiome of the Vector Lutzomyia longipalpisIs Essential for Survival of Leishmania infantum
- Author
-
Kelly, Patrick H., Bahr, Sarah M., Serafim, Tiago D., Ajami, Nadim J., Petrosino, Joseph F., Meneses, Claudio, Kirby, John R., Valenzuela, Jesus G., Kamhawi, Shaden, and Wilson, Mary E.
- Abstract
ABSTRACTThe vector-borne disease leishmaniasis, caused by Leishmaniaspecies protozoa, is transmitted to humans by phlebotomine sand flies. Development of Leishmaniato infective metacyclic promastigotes in the insect gut, a process termed metacyclogenesis, is an essential prerequisite for transmission. Based on the hypothesis that vector gut microbiota influence the development of virulent parasites, we sequenced midgut microbiomes in the sand fly Lutzomyia longipalpiswith or without Leishmania infantuminfection. Sucrose-fed sand flies contained a highly diverse, stable midgut microbiome. Blood feeding caused a decrease in microbial richness that eventually recovered. However, bacterial richness progressively decreased in L. infantum-infected sand flies. Acetobacteraceaespp. became dominant and numbers of Pseudomonadaceaespp. diminished coordinately as the parasite underwent metacyclogenesis and parasite numbers increased. Importantly, antibiotic-mediated perturbation of the midgut microbiome rendered sand flies unable to support parasite growth and metacyclogenesis. Together, these data suggest that the sand fly midgut microbiome is a critical factor for Leishmaniagrowth and differentiation to its infective state prior to disease transmission.IMPORTANCELeishmania infantum, a parasitic protozoan causing fatal visceral leishmaniasis, is transmitted to humans through the bite of the sand fly Lutzomyia longipalpis. Development of the parasite to its virulent metacyclic state occurs in the sand fly gut. In this study, the microbiota within the Lu. longipalpismidgut was delineated by 16S ribosomal DNA (rDNA) sequencing, revealing a highly diverse community composition that lost diversity as parasites developed to their metacyclic state and increased in abundance in infected flies. Perturbing sand fly gut microbiota with an antibiotic cocktail, which alone had no effect on either the parasite or the fly, arrested both the development of virulent parasites and parasite expansion. These findings indicate the importance of bacterial commensals within the insect vector for the development of virulent pathogens, and raise the possibility that impairing the microbial composition within the vector might represent a novel approach to control of vector-borne diseases.
- Published
- 2017
- Full Text
- View/download PDF
45. IgA-coated E. colienriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation
- Author
-
Viladomiu, Monica, Kivolowitz, Charles, Abdulhamid, Ahmed, Dogan, Belgin, Victorio, Daniel, Castellanos, Jim G., Woo, Viola, Teng, Fei, Tran, Nhan L., Sczesnak, Andrew, Chai, Christina, Kim, Myunghoo, Diehl, Gretchen E., Ajami, Nadim J., Petrosino, Joseph F., Zhou, Xi K., Schwartzman, Sergio, Mandl, Lisa A., Abramowitz, Meira, Jacob, Vinita, Bosworth, Brian, Steinlauf, Adam, Scherl, Ellen J., Wu, Hsin-Jung Joyce, Simpson, Kenneth W., and Longman, Randy S.
- Abstract
IgA-reactive E. coliin Crohn’s disease–associated spondyloarthritis link mucosal immunity and systemic inflammation.
- Published
- 2017
- Full Text
- View/download PDF
46. Effect of Sample Storage Conditions on Culture-Independent Bacterial Community Measures in Cystic Fibrosis Sputum Specimens
- Author
-
Zhao, Jiangchao, Li, Jun, Schloss, Patrick D., Kalikin, Linda M., Raymond, Tracy A., Petrosino, Joseph F., Young, Vincent B., and LiPuma, John J.
- Published
- 2011
- Full Text
- View/download PDF
47. Nasopharyngeal Proteobacteria are associated with viral etiology and acute wheezing in children with severe bronchiolitis.
- Author
-
Hyde, Embriette R., Petrosino, Joseph F., Piedra, Pedro A., Camargo, Carlos A., Espinola, Janice A., and Mansbach, Jonathan M.
- Published
- 2014
- Full Text
- View/download PDF
48. The Gut Microbiome Modulates Colon Tumorigenesis
- Author
-
Zackular, Joseph P., Baxter, Nielson T., Iverson, Kathryn D., Sadler, William D., Petrosino, Joseph F., Chen, Grace Y., and Schloss, Patrick D.
- Abstract
ABSTRACTRecent studies have shown that individuals with colorectal cancer have an altered gut microbiome compared to healthy controls. It remains unclear whether these differences are a response to tumorigenesis or actively drive tumorigenesis. To determine the role of the gut microbiome in the development of colorectal cancer, we characterized the gut microbiome in a murine model of inflammation-associated colorectal cancer that mirrors what is seen in humans. We followed the development of an abnormal microbial community structure associated with inflammation and tumorigenesis in the colon. Tumor-bearing mice showed enrichment in operational taxonomic units (OTUs) affiliated with members of the Bacteroides, Odoribacter, and Akkermansiagenera and decreases in OTUs affiliated with members of the Prevotellaceaeand Porphyromonadaceaefamilies. Conventionalization of germfree mice with microbiota from tumor-bearing mice significantly increased tumorigenesis in the colon compared to that for animals colonized with a healthy gut microbiome from untreated mice. Furthermore, at the end of the model, germfree mice colonized with microbiota from tumor-bearing mice harbored a higher relative abundance of populations associated with tumor formation in conventional animals. Manipulation of the gut microbiome with antibiotics resulted in a dramatic decrease in both the number and size of tumors. Our results demonstrate that changes in the gut microbiome associated with inflammation and tumorigenesis directly contribute to tumorigenesis and suggest that interventions affecting the composition of the microbiome may be a strategy to prevent the development of colon cancer.IMPORTANCEThe trillions of bacteria that live in the gut, known collectively as the gut microbiome, are important for normal functioning of the intestine. There is now growing evidence that disruptive changes in the gut microbiome are strongly associated with the development colorectal cancer. However, how the gut microbiome changes with time during tumorigenesis and whether these changes directly contribute to disease have not been determined. We demonstrate using a mouse model of inflammation-driven colon cancer that there are dramatic, continual alterations in the microbiome during the development of tumors, which are directly responsible for tumor development. Our results suggest that interventions that target these changes in the microbiome may be an effective strategy for preventing the development of colorectal cancer.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.