Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Gastro Hep Adv ; 2(8): 1082-1087, 2023.
Article in English | MEDLINE | ID: mdl-38088988

ABSTRACT

BACKGROUND AND AIMS: Informed consent should allow patients the appropriate time and conditions to make decisions about their care. However, consent is often obtained immediately prior to a colonoscopy. We conducted a quality improvement study to assess how a preprocedure consent video 2 days prior to an outpatient colonoscopy impacts patient satisfaction. METHODS: Patients undergoing outpatient colonoscopy at a large academic medical center opted in to a text messaging platform for procedural information. Our intervention was an informed consent video 2 days before the colonoscopy. Our primary outcome was a composite patient satisfaction score. Pre and postintervention scores were compared using ordinal or multinomial logistic models to calculate odds ratios (OR) or relative risk ratios and 95% confidence intervals (CI), adjusting for age and sex. RESULTS: 1109 and 1452 patients completed ≥1 survey question in the pre and postintervention phases, respectively. Overall patient satisfaction did not differ between groups [OR for a 1-point increment in satisfaction score between post- vs pre-intervention groups = 1.05; 95% CI: 0.90-1.22; P = .51]. Compared to preintervention, postintervention respondents were more likely to report higher satisfaction with time available to talk with their physician (OR of a 1-point increase in individual question response = 1.29; 95% CI: 1.09-1.54; P = .004). Compared to preintervention, more physicians in the postintervention phase rated satisfaction with consent process efficiency as "very satisfied" or "satisfied" (P < .001). CONCLUSION: An informed consent video prior to colonoscopy resulted in similar overall patient satisfaction. However, post-intervention, patients were more likely to report sufficient time to talk with their physician, and physicians reported higher satisfaction with consent efficiency.

3.
Immunity ; 56(2): 444-458.e5, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36720220

ABSTRACT

Crohn's disease (CD) is a chronic gastrointestinal disease that is increasing in prevalence worldwide. CD is multifactorial, involving the complex interplay of genetic, immune, and environmental factors, necessitating a system-level understanding of its etiology. To characterize cell-type-specific transcriptional heterogeneity in active CD, we profiled 720,633 cells from the terminal ileum and colon of 71 donors with varying inflammation status. Our integrated datasets revealed organ- and compartment-specific responses to acute and chronic inflammation; most immune changes were in cell composition, whereas transcriptional changes dominated among epithelial and stromal cells. These changes correlated with endoscopic inflammation, but small and large intestines exhibited distinct responses, which were particularly apparent when focusing on IBD risk genes. Finally, we mapped markers of disease-associated myofibroblast activation and identified CHMP1A, TBX3, and RNF168 as regulators of fibrotic complications. Altogether, our results provide a roadmap for understanding cell-type- and organ-specific differences in CD and potential directions for therapeutic development.


Subject(s)
Crohn Disease , Humans , Transcriptome , Colon , Ileum , Inflammation/genetics , Ubiquitin-Protein Ligases/genetics
4.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161260

ABSTRACT

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


Subject(s)
Gastrointestinal Microbiome , Health Status Disparities , Disease , Health , Humans , Mental Health , Publications
5.
Med ; 2(8): 892-894, 2021 08 13.
Article in English | MEDLINE | ID: mdl-35590164

ABSTRACT

Gut microbes sense various external cues, including those found in inflammatory conditions, yielding possibilities for connecting sensory circuits to effector pathways to engineer precise and scaled interventions to target pathway activity. Scott et al. combined directed evolution and a synthetic gene circuit in yeast to fine-tune sensing of extracellular ATP, showing promise as a live therapeutic in two murine models of chemical colitis and a murine model of antibody-mediated enteritis.1.


Subject(s)
Colitis , Enteritis , Microbiota , Animals , Colitis/chemically induced , Inflammation/drug therapy , Mice , Time
6.
Cell ; 178(5): 1041-1056, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442399

ABSTRACT

The current understanding of inflammatory bowel disease (IBD) pathogenesis implicates a complex interaction between host genetics, host immunity, microbiome, and environmental exposures. Mechanisms gleaned from genetics and molecular pathogenesis offer clues to the critical triggers of mucosal inflammation and guide the development of therapeutic interventions. A complex network of interactions between host genetic factors, microbes, and microbial metabolites governs intestinal homeostasis, making classification and mechanistic dissection of involved pathways challenging. In this Review, we discuss these challenges, areas of active translation, and opportunities for development of next-generation therapies.


Subject(s)
Inflammatory Bowel Diseases/pathology , Microbiota , Adaptive Immunity , Animals , Bacteria/genetics , Bacteria/metabolism , Biological Products/pharmacology , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Intestines/immunology , Intestines/microbiology , Microbiota/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Science ; 365(6449)2019 07 12.
Article in English | MEDLINE | ID: mdl-31296739

ABSTRACT

Characterizing the organization of the human gut microbiota is a formidable challenge given the number of possible interactions between its components. Using a statistical approach initially applied to financial markets, we measured temporally conserved covariance among bacterial taxa in the microbiota of healthy members of a Bangladeshi birth cohort sampled from 1 to 60 months of age. The results revealed an "ecogroup" of 15 covarying bacterial taxa that provide a concise description of microbiota development in healthy children from this and other low-income countries, and a means for monitoring community repair in undernourished children treated with therapeutic foods. Features of ecogroup population dynamics were recapitulated in gnotobiotic piglets as they transitioned from exclusive milk feeding to a fully weaned state consuming a representative Bangladeshi diet.


Subject(s)
Bacteria/classification , Child Nutrition Disorders/diet therapy , Child Nutrition Disorders/microbiology , Diet , Gastrointestinal Microbiome/physiology , Germ-Free Life , Animals , Bacteria/genetics , Bacteria/isolation & purification , Bangladesh , Bottle Feeding , Child, Preschool , Cohort Studies , Gastrointestinal Microbiome/genetics , Humans , Infant , Infant, Newborn , Models, Animal , Swine , Weaning
8.
Science ; 365(6449)2019 07 12.
Article in English | MEDLINE | ID: mdl-31296738

ABSTRACT

To examine the contributions of impaired gut microbial community development to childhood undernutrition, we combined metabolomic and proteomic analyses of plasma samples with metagenomic analyses of fecal samples to characterize the biological state of Bangladeshi children with severe acute malnutrition (SAM) as they transitioned, after standard treatment, to moderate acute malnutrition (MAM) with persistent microbiota immaturity. Host and microbial effects of microbiota-directed complementary food (MDCF) prototypes targeting weaning-phase bacterial taxa underrepresented in SAM and MAM microbiota were characterized in gnotobiotic mice and gnotobiotic piglets colonized with age- and growth-discriminatory bacteria. A randomized, double-blind controlled feeding study identified a lead MDCF that changes the abundances of targeted bacteria and increases plasma biomarkers and mediators of growth, bone formation, neurodevelopment, and immune function in children with MAM.


Subject(s)
Child Nutrition Disorders/diet therapy , Child Nutrition Disorders/microbiology , Gastrointestinal Microbiome , Germ-Free Life , Host Microbial Interactions , Infant Nutritional Physiological Phenomena , Animals , Bangladesh , Blood Proteins/analysis , Child Nutrition Disorders/metabolism , Child, Preschool , Humans , Infant
9.
Endocrinology ; 158(8): 2441-2452, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28637315

ABSTRACT

Prenatal undernutrition and low birth weight are associated with risk of type 2 diabetes and obesity. Prenatal caloric restriction results in low birth weight, glucose intolerance, obesity, and reduced plasma bile acids (BAs) in offspring mice. Because BAs can regulate systemic metabolism and glucose homeostasis, we hypothesized that BA supplementation could prevent diet-induced obesity and glucose intolerance in this model of developmental programming. Pregnant dams were food restricted by 50% from gestational days 12.5 to 18.5. Offspring of both undernourished (UN) and control (C) dams given unrestricted diets were weaned to high-fat diets with or without supplementation with 0.25% w/w ursodeoxycholic acid (UDCA), yielding four experimental groups: C, UN, C + UDCA, and UN + UDCA. Glucose homeostasis, BA composition, liver and intestinal gene expression, and microbiota composition were analyzed in the four groups. Although UDCA supplementation ameliorated diet-induced obesity in C mice, there was no effect in UN mice. UDCA similarly lowered fasting insulin, and improved glucose tolerance, pyruvate tolerance, and liver steatosis in C, but not UN, animals. BA composition differed significantly, and liver and ileal expression of genes involved in BA metabolism (Cyp7b1, Shp) were differentially induced by UDCA in C vs UN animals. Bacterial taxa in fecal microbiota correlated with treatment groups and metabolic parameters. In conclusion, prenatal undernutrition alters responsiveness to the metabolic benefits of BA supplementation, with resistance to the weight-lowering and insulin-sensitizing effects of UDCA supplementation. Our findings suggest that BA metabolism may be a previously unrecognized contributor to developmentally programmed diabetes risk.


Subject(s)
Bile Acids and Salts/pharmacology , Glucose/metabolism , Insulin Resistance/physiology , Malnutrition , Prenatal Nutritional Physiological Phenomena , Animals , Bile Acids and Salts/blood , Bile Acids and Salts/chemistry , Blood Glucose , Diet, High-Fat , Female , Male , Mice , Mice, Inbred ICR , Pregnancy , Prenatal Exposure Delayed Effects , Ursodeoxycholic Acid/administration & dosage , Ursodeoxycholic Acid/pharmacology
10.
Science ; 351(6275)2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26912898

ABSTRACT

Undernourished children exhibit impaired development of their gut microbiota. Transplanting microbiota from 6- and 18-month-old healthy or undernourished Malawian donors into young germ-free mice that were fed a Malawian diet revealed that immature microbiota from undernourished infants and children transmit impaired growth phenotypes. The representation of several age-discriminatory taxa in recipient animals correlated with lean body mass gain; liver, muscle, and brain metabolism; and bone morphology. Mice were cohoused shortly after receiving microbiota from healthy or severely stunted and underweight infants; age- and growth-discriminatory taxa from the microbiota of the former were able to invade that of the latter, which prevented growth impairments in recipient animals. Adding two invasive species, Ruminococcus gnavus and Clostridium symbiosum, to the microbiota from undernourished donors also ameliorated growth and metabolic abnormalities in recipient animals. These results provide evidence that microbiota immaturity is causally related to undernutrition and reveal potential therapeutic targets and agents.


Subject(s)
Bacteria/classification , Gastrointestinal Microbiome/physiology , Infant Nutrition Disorders/microbiology , Animals , Bifidobacterium/physiology , Body Weight , Bone Development , Clostridiales/physiology , Disease Models, Animal , Feces/microbiology , Femur/growth & development , Germ-Free Life , Humans , Infant , Infant Nutrition Disorders/metabolism , Malawi , Male , Mice , Mice, Inbred C57BL
11.
Cell ; 161(1): 36-48, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25815983

ABSTRACT

Microbiota assembly is perturbed in children with undernutrition, resulting in persistent microbiota immaturity that is not rescued by current nutritional interventions. Evidence is accumulating that this immaturity is causally related to the pathogenesis of undernutrition and its lingering sequelae. Preclinical models in which human gut communities are replicated in gnotobiotic mice have provided an opportunity to identify and predict the effects of different dietary ingredients on microbiota structure, expressed functions, and host biology. This capacity sets the stage for proof-of-concept tests designed to deliberately shape the developmental trajectory and configurations of microbiota in children representing different geographies, cultural traditions, and states of health. Developing these capabilities for microbial stewardship is timely given the global health burden of childhood undernutrition, the effects of changing eating practices brought about by globalization, and the realization that affordable nutritious foods need to be developed to enhance our capacity to cultivate healthier microbiota in populations at risk for poor nutrition.


Subject(s)
Child Development , Gastrointestinal Tract/microbiology , Microbiota , Animals , Child , Child Nutritional Physiological Phenomena , Female , Humans , Maternal-Fetal Exchange , Pregnancy
12.
Proc Natl Acad Sci U S A ; 112(7): 1929-36, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25691701

ABSTRACT

Unlike mammalian embryogenesis, which takes place in the relatively predictable and stable environment of the uterus, postnatal development can be affected by a multitude of highly variable environmental factors, including diet, exposure to noxious substances, and microorganisms. Microbial colonization of the intestine is thought to play a particularly important role in postnatal development of the gastrointestinal, metabolic, and immune systems. Major changes in environmental exposure occur right after birth, upon weaning, and during pubertal maturation into adulthood. These transitions include dramatic changes in intestinal contents and require appropriate adaptations to meet changes in functional demands. Here, we attempt to both characterize and provide mechanistic insights into postnatal intestinal ontogeny. We investigated changes in global intestinal gene expression through postnatal developmental transitions. We report profound alterations in small and large intestinal transcriptional programs that accompany both weaning and puberty in WT mice. Using myeloid differentiation factor 88 (MyD88)/TIR-domain-containing adapter-inducing interferon-ß (TRIF) double knockout littermates, we define the role of toll-like receptors (TLRs) and interleukin (IL)-1 receptor family member signaling in postnatal gene expression programs and select ontogeny-specific phenotypes, such as vascular and smooth muscle development and neonatal epithelial and mast cell homeostasis. Metaanalysis of the effect of the microbiota on intestinal gene expression allowed for mechanistic classification of developmentally regulated genes by TLR/IL-1R (TIR) signaling and/or indigenous microbes. We find that practically every aspect of intestinal physiology is affected by postnatal transitions. Developmental timing, microbial colonization, and TIR signaling seem to play distinct and specific roles in regulation of gene-expression programs throughout postnatal development.


Subject(s)
Gene-Environment Interaction , Intestines/growth & development , Animals , Computational Biology , Mice , Mice, Knockout , Receptors, Interleukin-1/genetics , Toll-Like Receptors/genetics
13.
Nat Commun ; 5: 5320, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25382143

ABSTRACT

Identifying the factors that influence the outcome of host-microbial interactions is critical to protecting biodiversity, minimizing agricultural losses and improving human health. A few genes that determine symbiosis or resistance to infectious disease have been identified in model species, but a comprehensive examination of how a host genotype influences the structure of its microbial community is lacking. Here we report the results of a field experiment with the model plant Arabidopsis thaliana to identify the fungi and bacteria that colonize its leaves and the host loci that influence the microbe numbers. The composition of this community differs among accessions of A. thaliana. Genome-wide association studies (GWAS) suggest that plant loci responsible for defense and cell wall integrity affect variation in this community. Furthermore, species richness in the bacterial community is shaped by host genetic variation, notably at loci that also influence the reproduction of viruses, trichome branching and morphogenesis.


Subject(s)
Arabidopsis/microbiology , Genome, Microbial/genetics , Genome-Wide Association Study , Plant Leaves/microbiology , Genes, Bacterial/genetics , Genes, Fungal/genetics , Genetic Loci/genetics , Genotype
14.
Nature ; 515(7527): 423-6, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25231861

ABSTRACT

Given the global burden of diarrhoeal diseases, it is important to understand how members of the gut microbiota affect the risk for, course of, and recovery from disease in children and adults. The acute, voluminous diarrhoea caused by Vibrio cholerae represents a dramatic example of enteropathogen invasion and gut microbial community disruption. Here we conduct a detailed time-series metagenomic study of faecal microbiota collected during the acute diarrhoeal and recovery phases of cholera in a cohort of Bangladeshi adults living in an area with a high burden of disease. We find that recovery is characterized by a pattern of accumulation of bacterial taxa that shows similarities to the pattern of assembly/maturation of the gut microbiota in healthy Bangladeshi children. To define the underlying mechanisms, we introduce into gnotobiotic mice an artificial community composed of human gut bacterial species that directly correlate with recovery from cholera in adults and are indicative of normal microbiota maturation in healthy Bangladeshi children. One of the species, Ruminococcus obeum, exhibits consistent increases in its relative abundance upon V. cholerae infection of the mice. Follow-up analyses, including mono- and co-colonization studies, establish that R. obeum restricts V. cholerae colonization, that R. obeum luxS (autoinducer-2 (AI-2) synthase) expression and AI-2 production increase significantly with V. cholerae invasion, and that R. obeum AI-2 causes quorum-sensing-mediated repression of several V. cholerae colonization factors. Co-colonization with V. cholerae mutants discloses that R. obeum AI-2 reduces Vibrio colonization/pathogenicity through a novel pathway that does not depend on the V. cholerae AI-2 sensor, LuxP. The approach described can be used to mine the gut microbiota of Bangladeshi or other populations for members that use autoinducers and/or other mechanisms to limit colonization with V. cholerae, or conceivably other enteropathogens.


Subject(s)
Cholera/microbiology , Intestines/microbiology , Microbiota/physiology , Ruminococcus/physiology , Vibrio cholerae/physiology , Vibrio cholerae/pathogenicity , Animals , Bangladesh , Child , Cohort Studies , Diarrhea/microbiology , Feces/microbiology , Gene Expression Regulation, Bacterial , Germ-Free Life , Health , Humans , Male , Metagenome/genetics , Mice , Microbiota/genetics , Quorum Sensing/physiology , Ruminococcus/isolation & purification , Vibrio cholerae/genetics , Vibrio cholerae/isolation & purification , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
15.
Nature ; 510(7505): 417-21, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24896187

ABSTRACT

Therapeutic food interventions have reduced mortality in children with severe acute malnutrition (SAM), but incomplete restoration of healthy growth remains a major problem. The relationships between the type of nutritional intervention, the gut microbiota, and therapeutic responses are unclear. In the current study, bacterial species whose proportional representation define a healthy gut microbiota as it assembles during the first two postnatal years were identified by applying a machine-learning-based approach to 16S ribosomal RNA data sets generated from monthly faecal samples obtained from birth onwards in a cohort of children living in an urban slum of Dhaka, Bangladesh, who exhibited consistently healthy growth. These age-discriminatory bacterial species were incorporated into a model that computes a 'relative microbiota maturity index' and 'microbiota-for-age Z-score' that compare postnatal assembly (defined here as maturation) of a child's faecal microbiota relative to healthy children of similar chronologic age. The model was applied to twins and triplets (to test for associations of these indices with genetic and environmental factors, including diarrhoea), children with SAM enrolled in a randomized trial of two food interventions, and children with moderate acute malnutrition. Our results indicate that SAM is associated with significant relative microbiota immaturity that is only partially ameliorated following two widely used nutritional interventions. Immaturity is also evident in less severe forms of malnutrition and correlates with anthropometric measurements. Microbiota maturity indices provide a microbial measure of human postnatal development, a way of classifying malnourished states, and a parameter for judging therapeutic efficacy. More prolonged interventions with existing or new therapeutic foods and/or addition of gut microbes may be needed to achieve enduring repair of gut microbiota immaturity in childhood malnutrition and improve clinical outcomes.


Subject(s)
Bacterial Physiological Phenomena , Biodiversity , Infant Nutrition Disorders/microbiology , Microbiota , Bacteria/classification , Bacteria/genetics , Bangladesh , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Humans , Infant , Infant Nutrition Disorders/diet therapy , Male , Models, Biological , Nutritional Status , RNA, Ribosomal, 16S/genetics
16.
Science ; 341(6141): 1237439, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23828941

ABSTRACT

A low-error 16S ribosomal RNA amplicon sequencing method, in combination with whole-genome sequencing of >500 cultured isolates, was used to characterize bacterial strain composition in the fecal microbiota of 37 U.S. adults sampled for up to 5 years. Microbiota stability followed a power-law function, which when extrapolated suggests that most strains in an individual are residents for decades. Shared strains were recovered from family members but not from unrelated individuals. Sampling of individuals who consumed a monotonous liquid diet for up to 32 weeks indicated that changes in strain composition were better predicted by changes in weight than by differences in sampling interval. This combination of stability and responsiveness to physiologic change confirms the potential of the gut microbiota as a diagnostic tool and therapeutic target.


Subject(s)
Gastrointestinal Tract/microbiology , Metagenome , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Body Composition , Caloric Restriction , Family , Feces/microbiology , Female , Genome, Bacterial/genetics , Genomic Instability , Humans , Male , Models, Biological , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Time Factors , Weight Loss , Young Adult
17.
Nat Methods ; 10(1): 57-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202435

ABSTRACT

High-throughput sequencing has revolutionized microbial ecology, but read quality remains a considerable barrier to accurate taxonomy assignment and α-diversity assessment for microbial communities. We demonstrate that high-quality read length and abundance are the primary factors differentiating correct from erroneous reads produced by Illumina GAIIx, HiSeq and MiSeq instruments. We present guidelines for user-defined quality-filtering strategies, enabling efficient extraction of high-quality data and facilitating interpretation of Illumina sequencing results.


Subject(s)
Biodiversity , High-Throughput Nucleotide Sequencing/methods , Quality Control , Sequence Analysis, DNA/methods , Humans
18.
Cytometry A ; 77(11): 1067-74, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20872885

ABSTRACT

Plasmodium falciparum genotyping has recently undergone a revolution, and genome-wide genotype datasets are now being collected for large numbers of parasite isolates. By contrast, phenotyping technologies have lagged behind, with few high throughput phenotyping platforms available. Invasion of human erythrocytes by Plasmodium falciparum is a phenotype of particular interest because of its central role in parasite development. Invasion is a variable phenotype influenced by natural genetic variation in both the parasite and host and is governed by multiple overlapping and in some instances redundant parasite-erythrocyte interactions. To facilitate the scale-up of erythrocyte invasion phenotyping, we have developed a novel platform based on two-color flow cytometry that distinguishes parasite invasion from parasite growth. Target cells that had one or more receptors removed using enzymatic treatment were prelabeled with intracellular dyes CFDA-SE or DDAO-SE, incubated with P. falciparum parasites, and parasites that had invaded either labeled or unlabeled cells were detected with fluorescent DNA-intercalating dyes Hoechst 33342 or SYBR Green I. Neither cell label interfered with erythrocyte invasion, and the combination of cell and parasite dyes recapitulated known invasion phenotypes for three standard laboratory strains. Three different dye combinations with minimal overlap have been validated, meaning the same assay can be adapted to instruments harboring several different combinations of laser lines. The assay is sensitive, operates in a 96-well format, and can be used to quantitate the impact of natural or experimental genetic variation on erythrocyte invasion efficiency.


Subject(s)
Erythrocytes/parasitology , Flow Cytometry/methods , Malaria, Falciparum/diagnosis , Plasmodium falciparum/isolation & purification , Acridines/chemistry , Cell Separation , Cells, Cultured , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Host-Parasite Interactions , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/physiology , Staining and Labeling , Succinimides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...