Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Pediatr Gastroenterol Nutr ; 76(5): 684-694, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36976575

ABSTRACT

Telehealth (TH) broadly encompasses remote activities of clinical care (telemedicine), provider and patient education, and general health services. The use of synchronous video for TH first occurred in 1964 and then catapulted to the forefront in 2020 during the coronavirus disease 2019 public health emergency. Due to the sudden need for increased TH utilization by nearly all health care providers at that time, TH became essential to clinical practice. However, its sustainable future is unclear in part given that best practices for TH in pediatric gastroenterology (GI), hepatology, and nutrition remain undefined and non-standardized. Key areas for review include historical perspective, general and subspeciality usage, health care disparities, quality of care and the provider-patient interaction, logistics and operations, licensure and liability, reimbursement and insurance coverage, research and quality improvement (QI) priorities, and future use of TH in pediatric GI with a call for advocacy. This position paper from the Telehealth Special Interest Group of North American Society of Gastroenterology, Hepatology and Nutrition provides recommendations for pediatric GI-focused TH best practices, reviews areas for research and QI growth, and presents advocacy opportunities.


Subject(s)
COVID-19 , Gastroenterology , Telemedicine , Child , Humans , Gastroenterology/education , Societies , North America , Societies, Medical
2.
JPGN Rep ; 3(2): e182, 2022 May.
Article in English | MEDLINE | ID: mdl-37168904

ABSTRACT

With the coronavirus disease 2019 public health emergency (PHE), telehealth (TH) became essential for continued delivery of care. Members of the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN) formed the Telehealth for Pediatric Gastrointestinal Care Now (TPGCN) working group and rapidly organized a telemedicine webinar to provide education and guidance. We aim to describe the webinar development and prospectively assess the effectiveness of this webinar-based educational intervention. Methods: NASPGHAN members who registered for the TPGCN webinar received pre- and post-webinar surveys. Outcome measures included a modified Telehealth Acceptance Model (TAM) survey and a Student Evaluation of Educational Quality (SEEQ) standardized instrument. Results: Seven hundred seventy-six NASPGHAN members participated in the webinar, 147 (33%) completed the pre-webinar survey; of these, 25 of 147 (17%) completed a post-webinar survey. Before the PHE, 50.3% of the pre-webinar survey participants had no TH knowledge. Webinar participants trended to have increased acceptance of TH for follow-up visits (pre-webinar, 68% versus post-webinar, 81%; P = 0.15) and chronic disease care (pre-webinar, 57% vs post-webinar, 81%; P = 0.01). The overall acceptance of TH as shown by TAM pre-webinar was 1.74 ± 0.8, which improved to 1.62 ± 0.8 post-webinar (lower scores indicate greater acceptance; P < 0.001). SEEQ results indicate that webinar material was understandable (post-webinar, 95%). Participants found breakout sessions informative and enjoyable (post-webinar, 91%). Conclusion: The TPGCN TH webinar was an effective educational intervention that fostered increased TH usage for follow-up and chronic care visits, improved TAM scores, and was well received by participants as seen by high SEEQ scores. Sustained and expanded pediatric gastrointestinal TH usage beyond the coronavirus disease 2019 PHE is expected.

3.
JPGN Rep ; 2(3): e099, 2021 Aug.
Article in English | MEDLINE | ID: mdl-37205940

ABSTRACT

Patients with very early onset inflammatory bowel disease (VEO-IBD) have a higher incidence of monogenic disease compared to older age groups. Age, alone, is a strong predictor for monogenic disease. We discuss a case of VEO-IBD in which the patient presented with severe and refractory enteropathy, leading to diagnosis of CTLA-4 haploinsufficiency. Genetic workup showed de novo heterozygous deletions of the CTLA-4 and ICOS genes. This case was unique, as the patient did not have the other manifestations commonly present with the disease. We advocate for early and routine genetic workup of VEO-IBD, as patients with monogenic IBD have high morbidity and mortality, if inadequately treated. Our patient did not respond to conventional treatment modalities and required targeted treatment with Abatacept, a CTLA-4 agonist.

4.
PLoS One ; 15(5): e0231891, 2020.
Article in English | MEDLINE | ID: mdl-32433684

ABSTRACT

OBJECTIVE: The SI gene encodes the sucrase-isomaltase enzyme, a disaccharidase expressed in the intestinal brush border. Hypomorphic SI variants cause recessive congenital sucrase-isomaltase deficiency (CSID) and related gastrointestinal (GI) symptoms. Among children presenting with chronic, idiopathic loose stools, we assessed the prevalence of CSID-associated SI variants relative to the general population and the relative GI symptom burden associated with SI genotype within the study population. METHODS: A prospective study conducted at 18 centers enrolled 308 non-Hispanic white children ≤18 years old who were experiencing chronic, idiopathic, loose stools at least once per week for >4 weeks. Data on demographics, GI symptoms, and genotyping for 37 SI hypomorphic variants were collected. Race/ethnicity-matched SI data from the Exome Aggregation Consortium (ExAC) database was used as the general population reference. RESULTS: Compared with the general population, the cumulative prevalence of hypomorphic SI variants was significantly higher in the study population (4.5% vs. 1.3%, P < .01; OR = 3.5 [95% CI: 6.1, 2.0]). Within the study population, children with a hypomorphic SI variant had a more severe GI symptom burden than those without, including: more frequent episodes of loose stools (P < .01), higher overall stool frequency (P < .01), looser stool form (P = .01) and increased flatulence (P = .02). CONCLUSION: Non-Hispanic white children with chronic idiopathic loose stools have a higher prevalence of CSID-associated hypomorphic SI variants than the general population. The GI symptom burden was greater among the study subjects with a hypomorphic SI variant than those without hypomorphic SI variants.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/pathology , Sucrase-Isomaltase Complex/deficiency , Sucrase-Isomaltase Complex/genetics , Adolescent , Carbohydrate Metabolism, Inborn Errors/epidemiology , Carbohydrate Metabolism, Inborn Errors/genetics , Child , Databases, Factual , Female , Genotype , Heterozygote , Humans , Male , Polymorphism, Single Nucleotide , Prevalence , Prospective Studies
5.
Inflamm Bowel Dis ; 24(6): 1321-1334, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29788359

ABSTRACT

Goal: The aim of this study was to investigate gene expression levels of proteins involved in sphingosine-1-phosphate (S1P) metabolism and signaling in a pediatric inflammatory bowel disease (IBD) patient population. Background: IBD is a debilitating disease affecting 0.4% of the US population. The incidence of IBD in childhood is rising. Identifying effective targeted therapies that can be used safely in young patients and developing tools for selecting specific candidates for targeted therapies are important goals. Clinical IBD trials now underway target S1PR1, a receptor for the pro-inflammatory sphingolipid S1P. However, circulating and tissue sphingolipid levels and S1P-related gene expression have not been characterized in pediatric IBD. Methods: Pediatric IBD patients and controls were recruited in a four-site study. Patients received a clinical score using PUCAI or PCDAI evaluation. Colon biopsies were collected during endoscopy. Gene expression was measured by qRT-PCR. Plasma and gut tissue sphingolipids were measured by LC-MS/MS. Results: Genes of S1P synthesis (SPHK1, SPHK2), degradation (SGPL1), and signaling (S1PR1, S1PR2, and S1PR4) were significantly upregulated in colon biopsies of IBD patients with moderate/severe symptoms compared with controls or patients in remission. Tissue ceramide, dihydroceramide, and ceramide-1-phosphate (C1P) levels were significantly elevated in IBD patients compared with controls. Conclusions: A signature of elevated S1P-related gene expression in colon tissues of pediatric IBD patients correlates with active disease and normalizes in remission. Biopsied gut tissue from symptomatic IBD patients contains high levels of pro-apoptotic and pro-inflammatory sphingolipids. A combined analysis of gut tissue sphingolipid profiles with this S1P-related gene signature may be useful for monitoring response to conventional therapy.


Subject(s)
Colon/metabolism , Gene Expression , Inflammatory Bowel Diseases/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Adolescent , Animals , Case-Control Studies , Ceramides/metabolism , Child , Child, Preschool , Chromatography, Liquid , Colon/pathology , Female , Humans , Infant , Inflammatory Bowel Diseases/genetics , Lysophospholipids/genetics , Male , Pilot Projects , Signal Transduction , Sphingosine/genetics , Sphingosine/metabolism , Tandem Mass Spectrometry , Young Adult
6.
Gastroenterology ; 149(3): 681-91.e10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26001928

ABSTRACT

BACKGROUND & AIMS: The mechanisms of tissue destruction during progression of celiac disease are poorly defined. It is not clear how tissue stress and adaptive immunity contribute to the activation of intraepithelial cytotoxic T cells and the development of villous atrophy. We analyzed epithelial cells and intraepithelial cytotoxic T cells in family members of patients with celiac disease, who were without any signs of adaptive antigluten immunity, and in potential celiac disease patients, who have antibodies against tissue transglutaminase 2 in the absence of villous atrophy. METHODS: We collected blood and intestinal biopsy specimens from 268 patients at tertiary medical centers in the United States and Italy from 2004 to 2012. All subjects had normal small intestinal histology. Study groups included healthy individuals with no family history of celiac disease or antibodies against tissue transglutaminase 2 (controls), healthy family members of patients with celiac disease, and potential celiac disease patients. Intraepithelial cytotoxic T cells were isolated and levels of inhibitory and activating natural killer (NK) cells were measured by flow cytometry. Levels of heat shock protein (HSP) and interleukin 15 were measured by immunohistochemistry, and ultrastructural alterations in intestinal epithelial cells (IECs) were assessed by electron microscopy. RESULTS: IECs from subjects with a family history of celiac disease, but not from subjects who already had immunity to gluten, expressed higher levels of HS27, HSP70, and interleukin-15 than controls; their IECs also had ultrastructural alterations. Intraepithelial cytotoxic T cells from relatives of patients with celiac disease expressed higher levels of activating NK receptors than cells from controls, although at lower levels than patients with active celiac disease, and without loss of inhibitory receptors for NK cells. Intraepithelial cytotoxic T cells from potential celiac disease patients failed to up-regulate activating NK receptors. CONCLUSIONS: A significant subset of healthy family members of patients with celiac disease with normal intestinal architecture had epithelial alterations, detectable by immunohistochemistry and electron microscopy. The adaptive immune response to gluten appears to act in synergy with epithelial stress to allow intraepithelial cytotoxic T cells to kill epithelial cells and induce villous atrophy in patients with active celiac disease.


Subject(s)
Adaptive Immunity , Celiac Disease/immunology , Cell Communication , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Intestine, Small/immunology , Stress, Physiological , T-Lymphocytes, Cytotoxic/immunology , Autoantibodies/blood , Case-Control Studies , Celiac Disease/blood , Celiac Disease/pathology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , GTP-Binding Proteins/immunology , HSP27 Heat-Shock Proteins/immunology , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Italy , Molecular Chaperones , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , Risk Factors , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure , Transglutaminases/immunology , United States
7.
J Pediatr Gastroenterol Nutr ; 51(1): 24-30, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20453678

ABSTRACT

BACKGROUND AND OBJECTIVES: : Irritable bowel syndrome (IBS) is a common problem in pediatrics, for which no safe and effective treatment is available. Probiotics have shown some promising results in adult studies, but no positive study has been published on pediatric age. We aimed at investigating the efficacy of VSL#3 in a population of children and teenagers affected by IBS, in a randomized, double-blind, placebo-controlled, crossover study conducted in 7 pediatric gastroenterology divisions. PATIENTS AND METHODS: : Children 4 to 18 years of age, meeting eligibility criteria, were enrolled. The patients were assessed by a questionnaire for a 2-week baseline period. They were then randomized to receive either VSL#3 or a placebo for 6 weeks, with controls every 2 weeks. At the end, after a "wash-out" period of 2 weeks, each patient was switched to the other group and followed for a further 6 weeks. RESULTS: : A total of 59 children completed the study. Although placebo was effective in some of the parameters and in as many as half of the patients, VSL#3 was significantly superior to it (P < 0.05) in the primary endpoint, the subjective assessment of relief of symptoms; as well as in 3 of 4 secondary endpoints: abdominal pain/discomfort (P < 0.05), abdominal bloating/gassiness (P < 0.05), and family assessment of life disruption (P < 0.01). No significant difference was found (P = 0.06) in the stool pattern. No untoward adverse effect was recorded in any of the patients. CONCLUSIONS: : VSL#3 is safe and more effective than placebo in ameliorating symptoms and improving the quality of life in children affected by IBS.


Subject(s)
Irritable Bowel Syndrome/drug therapy , Probiotics/therapeutic use , Abdominal Pain/drug therapy , Abdominal Pain/etiology , Adolescent , Child , Child, Preschool , Cross-Over Studies , Double-Blind Method , Family , Female , Humans , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/microbiology , Male , Surveys and Questionnaires , Treatment Outcome
9.
J Exp Med ; 206(3): 707-19, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19237603

ABSTRACT

IL-15 and NKG2D promote autoimmunity and celiac disease by arming cytotoxic T lymphocytes (CTLs) to cause tissue destruction. However, the downstream signaling events underlying these functional properties remain unclear. Here, we identify cytosolic phospholipase A(2) (cPLA(2)) as a central molecule in NKG2D-mediated cytolysis in CTLs. Furthermore, we report that NKG2D induces, upon recognition of MIC(+) target cells, the release of arachidonic acid (AA) by CTLs to promote tissue inflammation in association with target killing. Interestingly, IL-15, which licenses NKG2D-mediated lymphokine killer activity in CTLs, cooperates with NKG2D to induce cPLA(2) activation and AA release. Finally, cPLA(2) activation in intraepithelial CTLs of celiac patients provides an in vivo pathophysiological dimension to cPLA(2) activation in CTLs. These results reveal an unrecognized link between NKG2D and tissue inflammation, which may underlie the emerging role of NKG2D in various immunopathological conditions and define new therapeutic targets.


Subject(s)
Celiac Disease/enzymology , Celiac Disease/immunology , Interleukin-15/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phospholipases A2, Cytosolic/metabolism , T-Lymphocytes, Cytotoxic/enzymology , Arachidonic Acid/metabolism , Celiac Disease/pathology , Cell Degranulation , Cytotoxicity, Immunologic , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Models, Immunological , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/physiology , Up-Regulation
10.
Mol Diagn Ther ; 12(5): 289-98, 2008.
Article in English | MEDLINE | ID: mdl-18803427

ABSTRACT

Celiac disease is an autoimmune disorder occurring in genetically susceptible individuals, triggered by gluten and related prolamins. Well identified haplotypes in the human leukocyte antigen (HLA) class II region (either DQ2 [DQA*0501-DQB*0201] or DQ8 [DQA*0301-DQB1*0302]) confer a large part of the genetic susceptibility to celiac disease.Celiac disease originates as a result of a combined action involving both adaptive and innate immunity. The adaptive immune response to gluten has been well described, with the identification of specific peptide sequences demonstrating HLA-DQ2 or -DQ8 restrictive binding motifs across various gluten proteins. As for innate immunity, through specific natural killer receptors expressed on their surface, intra-epithelial lymphocytes recognize nonclassical major histocompatibility complex (MHC)-I molecules such as MICA, which are induced on the surface of enterocytes by stress and inflammation, and this interaction leads to their activation to become lymphokine-activated killing cells. Four possible presentations of celiac disease are recognized: (i) typical, characterized mostly by gastrointestinal signs and symptoms; (ii) atypical or extraintestinal, where gastrointestinal signs/symptoms are minimal or absent and a number of other manifestations are present; (iii) silent, where the small intestinal mucosa is damaged and celiac disease autoimmunity can be detected by serology, but there are no symptoms; and, finally, (iv) latent, where individuals possess genetic compatibility with celiac disease and may also show positive autoimmune serology, that have a normal mucosa morphology and may or may not be symptomatic.The diagnosis of celiac disease still rests on the demonstration of changes in the histology of the small intestinal mucosa. The classic celiac lesion occurs in the proximal small intestine with histologic changes of villous atrophy, crypt hyperplasia, and increased intraepithelial lymphocytosis. Currently, serological screening tests are utilized primarily to identify those individuals in need of a diagnostic endoscopic biopsy. The serum levels of immunoglobulin (Ig)A anti-tissue transglutaminase (or TG2) are the first choice in screening for celiac disease, displaying the highest levels of sensitivity (up to 98%) and specificity (around 96%). Anti-endomysium antibodies-IgA (EMA), on the other hand, have close to 100% specificity and a sensitivity of greater than 90%. The interplay between gliadin peptides and TG2 is responsible for the generation of novel antigenic epitopes, the TG2-generated deamidated gliadin peptides. Such peptides represent much more celiac disease-specific epitopes than native peptides, and deamidated gliadin antibodies (DGP) have shown promising results as serological markers for celiac disease. Serology has also been employed in monitoring the response to a gluten-free diet.Despite the gluten-free diet being so effective, there is a growing demand for alternative treatment options. In the future, new forms of treatment may include the use of gluten-degrading enzymes to be ingested with meals, the development of alternative, gluten-free grains by genetic modification, the use of substrates regulating intestinal permeability to prevent gluten entry across the epithelium, and, finally, the availability of different forms of immunotherapy.


Subject(s)
Celiac Disease/diagnosis , Celiac Disease/immunology , Autoantibodies/immunology , Celiac Disease/physiopathology , Celiac Disease/therapy , Diet Therapy , Glutens/immunology , Histocompatibility Antigens Class II , Humans , Immune System/physiology , Immune System/physiopathology , Immunotherapy , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Risk Assessment , Risk Factors
11.
Curr Opin Gastroenterol ; 24(6): 707-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19122520

ABSTRACT

PURPOSE OF REVIEW: Research in celiac disease is unraveling new findings at a high rate, and major advances seem to occur in all areas such as genetics, environmental factor, pathophysiology, and even prospective therapeutic implications. RECENT FINDINGS: New insight is being gained into the interplay between genetic and environmental factors causing celiac disease. In addition to the known human leukocyte antigen haplotypes, genome-wide studies have now identified additional susceptibility loci and the majority of newly discovered risk regions harbor genes controlling immune pathways. The mechanism of translocation of gliadin peptides across the intestinal barrier has been the subject of much investigation, and there is now evidence that the toxic 33-mer peptide can also be translocated transcellularly. As for the paracellular route, this appears to be enhanced by gliadin's stimulation of zonulin release. The growing role of the innate immunity is being recognized and the increased expression of some Toll-like receptors appears to delineate a new inherent defect in this branch of innate immunity. Finally, new perspectives are opening in the treatment of celiac disease based on new detoxified grains, enzymatic degradation of gluten, and prevention of its crossing the mucosal barrier. SUMMARY: The pace of new knowledge in this 'ancient' disease is very fast, and this review outlines the principal lines of such exciting developments.


Subject(s)
Autoimmunity/immunology , Celiac Disease , Diet, Gluten-Free/methods , Environmental Exposure/adverse effects , Genetic Predisposition to Disease , Immunologic Factors/therapeutic use , Animals , Celiac Disease/etiology , Celiac Disease/metabolism , Celiac Disease/therapy , Humans , Prognosis
12.
J Exp Med ; 203(5): 1343-55, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16682498

ABSTRACT

Celiac disease is an intestinal inflammatory disorder induced by dietary gluten in genetically susceptible individuals. The mechanisms underlying the massive expansion of interferon gamma-producing intraepithelial cytotoxic T lymphocytes (CTLs) and the destruction of the epithelial cells lining the small intestine of celiac patients have remained elusive. We report massive oligoclonal expansions of intraepithelial CTLs that exhibit a profound genetic reprogramming of natural killer (NK) functions. These CTLs aberrantly expressed cytolytic NK lineage receptors, such as NKG2C, NKp44, and NKp46, which associate with adaptor molecules bearing immunoreceptor tyrosine-based activation motifs and induce ZAP-70 phosphorylation, cytokine secretion, and proliferation independently of T cell receptor signaling. This NK transformation of CTLs may underlie both the self-perpetuating, gluten-independent tissue damage and the uncontrolled CTL expansion leading to malignant lymphomas in severe forms of celiac disease. Because similar changes were detected in a subset of CTLs from cytomegalovirus-seropositive patients, we suggest that a stepwise transformation of CTLs into NK-like cells may underlie immunopathology in various chronic infectious and inflammatory diseases.


Subject(s)
Celiac Disease/immunology , Cell Differentiation/immunology , Cell Proliferation , Killer Cells, Natural/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Base Sequence , Celiac Disease/complications , Celiac Disease/genetics , Celiac Disease/pathology , Chronic Disease , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Interferon-gamma/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Killer Cells, Natural/pathology , Lymphoma/etiology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/pathology , ZAP-70 Protein-Tyrosine Kinase/immunology
13.
J Pediatr Gastroenterol Nutr ; 42(5): 454-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16707966

ABSTRACT

The use of probiotics, once discussed primarily in the context of alternative medicine, is now entering mainstream medicine. However, only a few of the potential health benefits attributed to probiotics have been confirmed in well-designed, well-conducted, randomized, controlled trials. This is especially true in the pediatric population. We review here the available evidence on efficacy of probiotics in children in the prevention and treatment of gastrointestinal diseases. Although we restrict our analysis to the pediatric age, whenever potentially relevant information is available only from adult studies, they are examined as well. Probiotics have been most extensively studied in the treatment of diarrheal diseases, where their efficacy can be considered well established. Studies documenting effects in other childhood gastrointestinal illnesses are few, although some preliminary results are promising. Furthermore, only a limited number of probiotic strains have been tested, and, as the effects of different probiotic microorganisms are not equivalent, results cannot be generalized. Thus, at present, we have some positive certainties, lots of exciting promises and many unanswered questions.


Subject(s)
Gastrointestinal Diseases/therapy , Probiotics , Respiratory Tract Infections/therapy , Adult , Child , Clostridioides difficile , Constipation/therapy , Cross Infection , Dysentery/microbiology , Dysentery/therapy , Enterocolitis, Necrotizing/therapy , Food Hypersensitivity/therapy , Gastrointestinal Diseases/prevention & control , Helicobacter Infections/microbiology , Helicobacter Infections/therapy , Helicobacter pylori , Humans , Infant , Inflammatory Bowel Diseases/therapy , Irritable Bowel Syndrome/therapy , Meta-Analysis as Topic , Randomized Controlled Trials as Topic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...