Genetics of irritable bowel syndrome: shifting gear via biobank-scale studies
暂无分享,去创建一个
[1] A. Kurilshikov,et al. Environmental factors shaping the gut microbiome in a Dutch population , 2022, Nature.
[2] Pieter B. T. Neerincx,et al. Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch Microbiome Project , 2022, Nature Genetics.
[3] A. Kurilshikov,et al. Challenges and future directions for studying effects of host genetics on the gut microbiome , 2022, Nature Genetics.
[4] J. Rosmalen,et al. Cohort Profile Update: Lifelines, a three-generation cohort study and biobank , 2021, International journal of epidemiology.
[5] G. Abecasis,et al. GWAS of stool frequency provides insights into gastrointestinal motility and irritable bowel syndrome , 2021, Cell genomics.
[6] Wei Zhou,et al. Global Biobank Meta-analysis Initiative: powering genetic discovery across human diseases , 2021, medRxiv.
[7] M. Camilleri,et al. Bile Acid Diarrhea in Adults and Adolescents , 2021, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[8] D. Drossman,et al. Rome Foundation Clinical Diagnostic Criteria for Disorders of Gut-Brain Interaction (DGBI). , 2021, Gastroenterology.
[9] Alicia R. Martin,et al. Responsible use of polygenic risk scores in the clinic: potential benefits, risks and gaps , 2021, Nature Medicine.
[10] Laurent F. Thomas,et al. Genome-wide analysis of 53,400 people with irritable bowel syndrome highlights shared genetic pathways with mood and anxiety disorders , 2021, Nature Genetics.
[11] A. Franke,et al. Adult sucrase-isomaltase deficiency masquerading as IBS , 2021, Gut.
[12] Alicia R. Martin,et al. Genome-wide association studies , 2021, Nature Reviews Methods Primers.
[13] K. Kalari,et al. Multi-omics analyses show disease, diet, and transcriptome interactions with the virome. , 2021, Gastroenterology.
[14] M. D’Amato,et al. Rare hypomorphic sucrase isomaltase variants in relation to IBS risk in UK Biobank. , 2021, Gastroenterology.
[15] R. Spiller,et al. The alternative serotonin transporter promoter P2 impacts gene function in females with irritable bowel syndrome , 2021, Journal of cellular and molecular medicine.
[16] N. Sousa,et al. Sorting Out Sorting Nexins Functions in the Nervous System in Health and Disease , 2021, Molecular Neurobiology.
[17] A. Kurilshikov,et al. Role of the gut microbiome in mediating lactose intolerance symptoms , 2021, Gut.
[18] M. Camilleri. Diagnosis and Treatment of Irritable Bowel Syndrome: A Review. , 2021, JAMA.
[19] P. Visscher,et al. GWAS of peptic ulcer disease implicates Helicobacter pylori infection, other gastrointestinal disorders and depression , 2021, Nature Communications.
[20] M. Camilleri. Gastrointestinal motility disorders in neurologic disease. , 2021, The Journal of clinical investigation.
[21] Judy H. Cho,et al. Common and rare variant prediction and penetrance of IBD in a large, multi-ethnic, health system-based biobank cohort. , 2020, Gastroenterology.
[22] M. Neurath,et al. All are Equal, Some are More Equal: Targeting IL 12 and 23 in IBD – A Clinical Perspective , 2020, ImmunoTargets and therapy.
[23] V. Salomaa,et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort , 2020, Nature Genetics.
[24] A. Hong,et al. SCF-FBXO24 regulates cell proliferation by mediating ubiquitination and degradation of PRMT6. , 2020, Biochemical and biophysical research communications.
[25] A. Regev,et al. The Human and Mouse Enteric Nervous System at Single-Cell Resolution , 2020, Cell.
[26] A. Peters,et al. Genome-wide association study in 8,956 German individuals identifies influence of ABO histo-blood groups on gut microbiome , 2020, Nature Genetics.
[27] Shiraz A. Shah,et al. Large-scale association analyses identify host factors influencing human gut microbiome composition , 2020, Nature Genetics.
[28] J. Raes,et al. Genome-wide associations of human gut microbiome variation and implications for causal inference analyses , 2020, Nature Microbiology.
[29] M. D’Amato,et al. Hypomorphic SI genetic variants are associated with childhood chronic loose stools , 2020, PloS one.
[30] M. Camilleri,et al. Actionable biomarkers: the key to resolving disorders of gastrointestinal function , 2020, Gut.
[31] M. Hoaglin,et al. Gut microbiome composition and risk factors in a large cross-sectional IBS cohort , 2020, BMJ open gastroenterology.
[32] P. Moayyedi,et al. Evidence-based and mechanistic insights into exclusion diets for IBS , 2020, Nature Reviews Gastroenterology & Hepatology.
[33] Padhmanand Sudhakar,et al. Big data in IBD: big progress for clinical practice , 2020, Gut.
[34] B. Tang. Syntaxin 16’s Newly Deciphered Roles in Autophagy , 2019, Cells.
[35] X. de la Cruz,et al. PHF2 histone demethylase prevents DNA damage and genome instability by controlling cell cycle progression of neural progenitors , 2019, Proceedings of the National Academy of Sciences.
[36] Christopher M. DeBoever,et al. Assessing digital phenotyping to enhance genetic studies of human diseases , 2019, bioRxiv.
[37] I. Hall,et al. Abnormalities of mucosal serotonin metabolism and 5‐HT3 receptor subunit 3C polymorphism in irritable bowel syndrome with diarrhoea predict responsiveness to ondansetron , 2019, Alimentary pharmacology & therapeutics.
[38] Aviv Regev,et al. Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis , 2019, Cell.
[39] B. Ohlsson,et al. A Dietary Intervention with Reduction of Starch and Sucrose Leads to Reduced Gastrointestinal and Extra-Intestinal Symptoms in IBS Patients , 2019, Nutrients.
[40] M. Surette,et al. Gut Microbiota in Patients With Irritable Bowel Syndrome-A Systematic Review. , 2019, Gastroenterology.
[41] Nicholas D. Youngblut,et al. Human salivary amylase gene copy number impacts oral and gut microbiomes , 2018, bioRxiv.
[42] Scott M. Williams,et al. The Missing Diversity in Human Genetic Studies , 2019, Cell.
[43] M. McCarthy,et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases , 2019, Nature Genetics.
[44] W. Chey,et al. Reduced efficacy of low FODMAPs diet in patients with IBS-D carrying sucrase-isomaltase (SI) hypomorphic variants , 2019, Gut.
[45] O. Andreassen,et al. A global overview of pleiotropy and genetic architecture in complex traits , 2019, Nature Genetics.
[46] Morris A. Swertz,et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome , 2018, Science Translational Medicine.
[47] M. Bonder,et al. Analysis of 1135 gut metagenomes identifies sex-specific resistome profiles , 2018, Gut microbes.
[48] Nayoung Kim,et al. Sex-Gender Differences in Irritable Bowel Syndrome , 2018, Journal of neurogastroenterology and motility.
[49] P. Donnelly,et al. The UK Biobank resource with deep phenotyping and genomic data , 2018, Nature.
[50] Sumei Liu. Neurotrophic factors in enteric physiology and pathophysiology , 2018, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[51] H. Törnblom,et al. Work Productivity and Activity Impairment in Irritable Bowel Syndrome (IBS): A Multifaceted Problem , 2018, The American Journal of Gastroenterology.
[52] Day Fr,et al. Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk , 2018, Yearbook of Paediatric Endocrinology.
[53] A. Zhernakova,et al. A GWAS meta‐analysis from 5 population‐based cohorts implicates ion channel genes in the pathogenesis of irritable bowel syndrome , 2018, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[54] Zachary F. Gerring,et al. GWAS of lifetime cannabis use reveals new risk loci, genetic overlap with psychiatric traits, and a causal effect of schizophrenia liability , 2018, Nature Neuroscience.
[55] A. Zhernakova,et al. Female-Specific Association Between Variants on Chromosome 9 and Self-Reported Diagnosis of Irritable Bowel Syndrome. , 2018, Gastroenterology.
[56] J. Fins. Family Portrait , 2018, Narrative inquiry in bioethics.
[57] S. Cole,et al. Radical and lunatic fringes modulate notch ligands to support mammalian intestinal homeostasis , 2018, eLife.
[58] D. Tester,et al. Irritable bowel syndrome patients have SCN5A channelopathies that lead to decreased NaV1.5 current and mechanosensitivity. , 2018, American journal of physiology. Gastrointestinal and liver physiology.
[59] D. Posthuma,et al. Item-level analyses reveal genetic heterogeneity in neuroticism , 2018, Nature Communications.
[60] A. Zhernakova,et al. The importance of cohort studies in the post-GWAS era , 2018, Nature Genetics.
[61] A. Kurilshikov,et al. Environment dominates over host genetics in shaping human gut microbiota , 2018, Nature.
[62] W. Chey,et al. Increased Prevalence of Rare Sucrase-isomaltase (SI) Pathogenic Variants in Irritable Bowel Syndrome Patients , 2018, Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association.
[63] Lin Chang,et al. FUNCTIONAL BOWEL DISORDERS , 2018, The American Journal of Gastroenterology.
[64] W. Lieb,et al. Sucrase-isomaltase 15Phe IBS risk variant in relation to dietary carbohydrates and faecal microbiota composition , 2018, Gut.
[65] J. Kaunitz,et al. Free fatty acid receptor 3 activation suppresses neurogenic motility in rat proximal colon , 2018, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[66] M. Delgado-Rodríguez,et al. Systematic review and meta-analysis. , 2017, Medicina intensiva.
[67] H. Dweep,et al. miR-16 and miR-103 impact 5-HT4 receptor signalling and correlate with symptom profile in irritable bowel syndrome , 2017, Scientific Reports.
[68] W. D. de Jonge,et al. Intestinal Fungal Dysbiosis Is Associated With Visceral Hypersensitivity in Patients With Irritable Bowel Syndrome and Rats. , 2017, Gastroenterology.
[69] P. Dupont,et al. Altered brain and gut responses to corticotropin-releasing hormone (CRH) in patients with irritable bowel syndrome , 2017, Scientific Reports.
[70] D. D. de Quervain,et al. The NCAM1 gene set is linked to depressive symptoms and their brain structural correlates in healthy individuals. , 2017, Journal of psychiatric research.
[71] David Julius,et al. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways , 2017, Cell.
[72] S. Slaugenhaupt,et al. Familial dysautonomia: History, genotype, phenotype and translational research , 2017, Progress in Neurobiology.
[73] Maristella,et al. Genomic analyses identify hundreds of variants associated with age at menarche and 1 support a role for puberty timing in cancer risk 2 , 2017 .
[74] M. Amiri,et al. Molecular pathogenicity of novel sucrase-isomaltase mutations found in congenital sucrase-isomaltase deficiency patients. , 2017, Biochimica et biophysica acta. Molecular basis of disease.
[75] A. Auton,et al. Replication and characterization of CADM2 and MSRA genes on human behavior , 2017, bioRxiv.
[76] R. Carson,et al. Health-related quality of life, work productivity, and indirect costs among patients with irritable bowel syndrome with diarrhea , 2017, Health and Quality of Life Outcomes.
[77] M. D’Amato,et al. TRPM8 polymorphisms associated with increased risk of IBS-C and IBS-M , 2016, Gut.
[78] M. Camilleri,et al. Functional variants in the sucrase–isomaltase gene associate with increased risk of irritable bowel syndrome , 2016, Gut.
[79] J. Christianson,et al. Ion channels, ion channel receptors, and visceral hypersensitivity in irritable bowel syndrome , 2016, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[80] Jessica L. Gören. THE PHARMACODYNAMICS OF MEDICATIONS USED IN PSYCHIATRIC DISORDERS , 2016 .
[81] A. Paterson,et al. Association of host genome with intestinal microbial composition in a large healthy cohort , 2016, Nature Genetics.
[82] J. Crispino,et al. Novel function of FAXDC2 in megakaryopoiesis , 2016, Blood Cancer Journal.
[83] M. Bonder,et al. A GWAS meta-analysis suggests roles for xenobiotic metabolism and ion channel activity in the biology of stool frequency , 2016, Gut.
[84] N. Israsena,et al. Stem Cells in the Intestine: Possible Roles in Pathogenesis of Irritable Bowel Syndrome , 2016, Journal of neurogastroenterology and motility.
[85] David C. Wilson,et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease , 2016, Nature Genetics.
[86] Emily R. Davenport,et al. Genetic Determinants of the Gut Microbiome in UK Twins. , 2016, Cell host & microbe.
[87] D. Drossman,et al. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. , 2016, Gastroenterology.
[88] D. Drossman. Functional Gastrointestinal Disorders: History, Pathophysiology, Clinical Features and Rome IV. , 2016, Gastroenterology.
[89] Morris A. Swertz,et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity , 2016, Science.
[90] A. Franke,et al. Stool frequency is associated with gut microbiota composition , 2016, Gut.
[91] J. Sundquist,et al. Familial risks of glomerulonephritis – a nationwide family study in Sweden , 2016, Annals of medicine.
[92] Blair H. Smith,et al. GWAS for executive function and processing speed suggests involvement of the CADM2 gene , 2016, Molecular Psychiatry.
[93] M. Amiri,et al. The multiple roles of sucrase-isomaltase in the intestinal physiology , 2016, Molecular and Cellular Pediatrics.
[94] M. Bonder,et al. Gut microbiota composition associated with stool consistency , 2015, Gut.
[95] S. Brooks,et al. Sex differences in gut fermentation and immune parameters in rats fed an oligofructose-supplemented diet , 2015, Biology of Sex Differences.
[96] B. Gomis. A Multifaceted Problem , 2015 .
[97] C. Wijmenga,et al. Cohort Profile Cohort Profile : LifeLines , a three-generation cohort study and biobank , 2015 .
[98] Judy H. Cho,et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations , 2015, Nature Genetics.
[99] L. Öhman,et al. Gut Microbiota as Potential Orchestrators of Irritable Bowel Syndrome , 2015, Gut and liver.
[100] R. Anney,et al. Common polygenic variation in coeliac disease and confirmation of ZNF335 and NIFA as disease susceptibility loci , 2015, European Journal of Human Genetics.
[101] Y. Taché,et al. Role of Corticotropin-releasing Factor Signaling in Stress-related Alterations of Colonic Motility and Hyperalgesia , 2015, Journal of neurogastroenterology and motility.
[102] E. Stoeckli,et al. The SynCAM synaptic cell adhesion molecules are involved in sensory axon pathfinding by regulating axon–axon contacts , 2014, Journal of Cell Science.
[103] X. Zuo,et al. Brain‐derived neurotrophic factor accelerates gut motility in slow‐transit constipation , 2014, Acta physiologica.
[104] Boris Yamrom,et al. The contribution of de novo coding mutations to autism spectrum disorder , 2014, Nature.
[105] J. Kere,et al. NPSR1 polymorphisms influence recurrent abdominal pain in children: a population‐based study , 2014, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[106] K. Browning,et al. Central nervous system control of gastrointestinal motility and secretion and modulation of gastrointestinal functions. , 2014, Comprehensive Physiology.
[107] S. Ward,et al. Interstitial cells: regulators of smooth muscle function. , 2014, Physiological reviews.
[108] W. Tourtellotte,et al. A neuron autonomous role for the familial dysautonomia gene ELP1 in sympathetic and sensory target tissue innervation , 2014, Development.
[109] M. Meleine,et al. Gender-related differences in irritable bowel syndrome: potential mechanisms of sex hormones. , 2014, World journal of gastroenterology.
[110] M. Gazouli,et al. Loss-of-function of the voltage-gated sodium channel NaV1.5 (channelopathies) in patients with irritable bowel syndrome. , 2014, Gastroenterology.
[111] R. Scott,et al. Genome-Wide Association Study Identifies Two Novel Genomic Regions in Irritable Bowel Syndrome , 2014, The American Journal of Gastroenterology.
[112] J. Koch,et al. BAG-6, a jack of all trades in health and disease , 2014, Cellular and Molecular Life Sciences.
[113] J. Sundquist,et al. Risk of irritable bowel syndrome in first-degree, second-degree and thirddegree relatives of affected individuals: a nationwide family study in Sweden , 2014, Gut.
[114] L. Avery,et al. Gastrointestinal symptoms before and during menses in healthy women , 2014, BMC Women's Health.
[115] Lei Shi. Dock protein family in brain development and neurological disease , 2013, Communicative & integrative biology.
[116] R. Khanna,et al. Peppermint Oil for the Treatment of Irritable Bowel Syndrome: A Systematic Review and Meta-analysis , 2013, Journal of clinical gastroenterology.
[117] M. Daly,et al. Exploring the genetics of irritable bowel syndrome: a GWA study in the general population and replication in multinational case-control cohorts , 2013, Gut.
[118] B. Naliboff,et al. Type, rather than number, of mental and physical comorbidities increases the severity of symptoms in patients with irritable bowel syndrome. , 2013, Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association.
[119] M. D’Amato. Genes and functional GI disorders: from casual to causal relationship , 2013, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[120] J. Hoffman,et al. Serotonin signalling in the gut—functions, dysfunctions and therapeutic targets , 2013, Nature Reviews Gastroenterology &Hepatology.
[121] T. Schwartz,et al. GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes. , 2013, Endocrinology.
[122] H. Friess,et al. Neural plasticity in the gastrointestinal tract: chronic inflammation, neurotrophic signals, and hypersensitivity , 2013, Acta Neuropathologica.
[123] P. Moayyedi,et al. Peripheral mechanisms in irritable bowel syndrome. , 2013, The New England journal of medicine.
[124] H. Kawahara,et al. BAG6/BAT3: emerging roles in quality control for nascent polypeptides. , 2013, Journal of biochemistry.
[125] David C. Wilson,et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease , 2012, Nature.
[126] A. Zinsmeister,et al. Pharmacodynamic and Clinical Endpoints for Functional Colonic Disorders: Statistical Considerations , 2012, Digestive Diseases and Sciences.
[127] Michael Camilleri,et al. Irritable bowel syndrome: methods, mechanisms, and pathophysiology. Genetic epidemiology and pharmacogenetics in irritable bowel syndrome. , 2012, American journal of physiology. Gastrointestinal and liver physiology.
[128] G. Breen,et al. A Polymorphism Associated with Depressive Disorders Differentially Regulates Brain Derived Neurotrophic Factor Promoter IV Activity , 2012, Biological Psychiatry.
[129] C. Wahlestedt,et al. Natural Antisense Inhibition Results in Transcriptional De-Repression and Gene Upregulation , 2012, Nature biotechnology.
[130] P. Visscher,et al. Five years of GWAS discovery. , 2012, American journal of human genetics.
[131] M. Heitkemper,et al. Genetics and Gastrointestinal Symptoms , 2011, Annual Review of Nursing Research.
[132] Sarah Edkins,et al. Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease , 2011, Nature Genetics.
[133] J. Miquel,et al. The European lactase persistence genotype determines the lactase persistence state and correlates with gastrointestinal symptoms in the Hispanic and Amerindian Chilean population: a case–control and population-based study , 2011, BMJ Open.
[134] A. Zinsmeister,et al. A Klothoβ variant mediates protein stability and associates with colon transit in irritable bowel syndrome with diarrhea. , 2011, Gastroenterology.
[135] J. Bueller,et al. The HTR3A polymorphism c. -42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. , 2011, Gastroenterology.
[136] Yuri A Saito. The role of genetics in IBS. , 2011, Gastroenterology clinics of North America.
[137] G. Ast,et al. IKAP/Elp1 involvement in cytoskeleton regulation and implication for familial dysautonomia. , 2011, Human molecular genetics.
[138] J. Tack,et al. TRP channels in neurogastroenterology: opportunities for therapeutic intervention , 2011, British journal of pharmacology.
[139] Christian Gieger,et al. Thirty new loci for age at menarche identified by a meta-analysis of genome-wide association studies , 2010, Nature Genetics.
[140] Min Goo Lee,et al. Syntaxin 16 Binds to Cystic Fibrosis Transmembrane Conductance Regulator and Regulates Its Membrane Trafficking in Epithelial Cells* , 2010, The Journal of Biological Chemistry.
[141] P. Whorwell,et al. 5-HTTLPR and STin2 polymorphisms in the serotonin transporter gene and irritable bowel syndrome: effect of bowel habit and sex , 2010, European journal of gastroenterology & hepatology.
[142] P. de Coppet,et al. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. , 2010, Gastroenterology.
[143] P. Deloukas,et al. Multiple common variants for celiac disease influencing immune gene expression , 2010, Nature Genetics.
[144] L. Öhman,et al. Pathogenesis of IBS: role of inflammation, immunity and neuroimmune interactions , 2010, Nature Reviews Gastroenterology &Hepatology.
[145] Li-Huei Tsai,et al. Cyclin-dependent kinases: a family portrait , 2009, Nature Cell Biology.
[146] A. Cole. PCTK Proteins: The Forgotten Brain Kinases? , 2009, Neurosignals.
[147] A. Baird,et al. Estrogen and its role in gastrointestinal health and disease , 2009, International Journal of Colorectal Disease.
[148] M. Camilleri. Genetics and Irritable Bowel Syndrome: From Genomics to Intermediate Phenotype and Pharmacogenetics , 2009, Digestive Diseases and Sciences.
[149] I. Kato,et al. Expression of short-chain fatty acid receptor GPR41 in the human colon. , 2009, Biomedical research.
[150] H. Katoh,et al. Regulation of dendrite growth by the Cdc42 activator Zizimin1/Dock9 in hippocampal neurons , 2009, Journal of neuroscience research.
[151] Klaus-Peter Zimmer,et al. Compound heterozygous mutations affect protein folding and function in patients with congenital sucrase-isomaltase deficiency. , 2009, Gastroenterology.
[152] Y. Taché,et al. Neuroendocrine control of the gut during stress: corticotropin-releasing factor signaling pathways in the spotlight. , 2009, Annual review of physiology.
[153] Takashi,et al. RESOLUTION , 2009, Bring Now the Angels.
[154] Cisca Wijmenga,et al. Shared and distinct genetic variants in type 1 diabetes and celiac disease. , 2008, The New England journal of medicine.
[155] Masashi Yanagisawa,et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41 , 2008, Proceedings of the National Academy of Sciences.
[156] P. Whorwell,et al. First evidence for an association of a functional variant in the microRNA-510 target site of the serotonin receptor-type 3E gene with diarrhea predominant irritable bowel syndrome. , 2008, Human molecular genetics.
[157] P. Maness,et al. L1 and NCAM adhesion molecules as signaling coreceptors in neuronal migration and process outgrowth , 2008, Current Opinion in Neurobiology.
[158] M. Camilleri,et al. An exploratory study of the association of adrenergic and serotonergic genotype and gastrointestinal motor functions , 2008, Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society.
[159] L. Kassem,et al. Sequence variation in DOCK9 and heterogeneity in bipolar disorder , 2007, Psychiatric genetics.
[160] C. Lindgren,et al. Neuropeptide s receptor 1 gene polymorphism is associated with susceptibility to inflammatory bowel disease. , 2007, Gastroenterology.
[161] Y. Nie,et al. The Association of Serotonin Transporter Genetic Polymorphisms and Irritable Bowel Syndrome and Its Influence on Tegaserod Treatment in Chinese Patients , 2007, Digestive Diseases and Sciences.
[162] B. Bonaz,et al. Corticotropin-releasing factor receptors and stress-related alterations of gut motor function. , 2007, The Journal of clinical investigation.
[163] I. Rooman,et al. Expression of the Notch signaling pathway and effect on exocrine cell proliferation in adult rat pancreas. , 2006, The American journal of pathology.
[164] A. Zinsmeister,et al. Gastrointestinal Symptoms in Families of Patients with an SCN5A-Encoded Cardiac Channelopathy: Evidence of an Intestinal Channelopathy , 2006, The American Journal of Gastroenterology.
[165] J. Grider,et al. Brain-derived neurotrophic factor augments peristalsis by augmenting 5-HT and calcitonin gene-related peptide release. , 2006, Gastroenterology.
[166] D. Drossman,et al. Psychiatric and psychological dysfunction in irritable bowel syndrome and the role of psychological treatments. , 2005, Gastroenterology clinics of North America.
[167] L. Chang,et al. Sex specific alterations in autonomic function among patients with irritable bowel syndrome , 2005, Gut.
[168] J. Ott,et al. Complement Factor H Polymorphism in Age-Related Macular Degeneration , 2005, Science.
[169] S. Moore,et al. Acetylcholinesterase in Hirschsprung’s disease , 2005, Pediatric Surgery International.
[170] M. Hongo,et al. Effect of a corticotropin releasing hormone receptor antagonist on colonic sensory and motor function in patients with irritable bowel syndrome , 2004, Gut.
[171] A. Zinsmeister,et al. Association of distinct α2 adrenoceptor and serotonin transporter polymorphisms with constipation and somatic symptoms in functional gastrointestinal disorders , 2004, Gut.
[172] L. Olds,et al. Lactase persistence DNA variant enhances lactase promoter activity in vitro: functional role as a cis regulatory element. , 2003, Human molecular genetics.
[173] Lei Dong,et al. Effects of neurotrophins on gastrointestinal myoelectric activities of rats. , 2003, World journal of gastroenterology.
[174] M. Heitkemper,et al. Impact of Sex and Gender on Irritable Bowel Syndrome , 2003, Biological research for nursing.
[175] I. Gottesman,et al. The endophenotype concept in psychiatry: etymology and strategic intentions. , 2003, The American journal of psychiatry.
[176] M. Camilleri,et al. Serotonin-transporter polymorphism pharmacogenetics in diarrhea-predominant irritable bowel syndrome. , 2002, Gastroenterology.
[177] W. Whitehead,et al. Systematic review of the comorbidity of irritable bowel syndrome with other disorders: what are the causes and implications? , 2002, Gastroenterology.
[178] P. Whorwell,et al. The menstrual cycle affects rectal sensitivity in patients with irritable bowel syndrome but not healthy volunteers , 2002, Gut.
[179] M. Camilleri,et al. Gender-related differences in slowing colonic transit by a 5-HT3 antagonist in subjects with diarrhea-predominant irritable bowel syndrome , 2001, American Journal of Gastroenterology.
[180] H. Soreq,et al. Acetylcholinesterase — new roles for an old actor , 2001, Nature Reviews Neuroscience.
[181] D. DeMets,et al. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework , 2001, Clinical pharmacology and therapeutics.
[182] M. Camilleri,et al. Recombinant human neurotrophic factors accelerate colonic transit and relieve constipation in humans. , 2000, Gastroenterology.
[183] D. Drossman. Do psychosocial factors define symptom severity and patient status in irritable bowel syndrome? , 1999, The American journal of medicine.
[184] F. Gleeson,et al. Onset of sucrase-isomaltase deficiency in late adulthood , 1999, American Journal of Gastroenterology.
[185] M. Hongo,et al. Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal controls and patients with irritable bowel syndrome , 1998, Gut.
[186] S. Lewis,et al. Stool form scale as a useful guide to intestinal transit time. , 1997, Scandinavian journal of gastroenterology.
[187] S. Phillips,et al. How well does stool form reflect colonic transit? , 1996, Gut.
[188] S. Leeder,et al. A population based study , 1993, The Medical journal of Australia.
[189] L. Belluscio,et al. NT-3, BDNF, and NGF in the developing rat nervous system: Parallel as well as reciprocal patterns of expression , 1990, Neuron.
[190] W. Whitehead,et al. Evidence for exacerbation of irritable bowel syndrome during menses. , 1990, Gastroenterology.
[191] Y. Taché,et al. Central nervous system action of peptides to influence gastrointestinal motor function. , 1990, Gastroenterology.
[192] G. Sturniolo,et al. [Irritable bowel syndrome]. , 1988, Giornale di clinica medica.
[193] H. Preiser,et al. Sucrase-isomaltase (palatinase) deficiency diagnosed during adulthood , 1980, Digestive Diseases and Sciences.
[194] Adam L. Haber,et al. Cellular and inter-cellular rewiring of the human colon during ulcerative colitis , 2019 .
[195] A. Zinsmeister,et al. Neuropeptide S receptor induces neuropeptide expression and associates with intermediate phenotypes of functional gastrointestinal disorders. , 2010, Gastroenterology.
[196] Hinrich W. H. Göhlmann,et al. Alterations in expression of p11 and SERT in mucosal biopsy specimens of patients with irritable bowel syndrome. , 2007, Gastroenterology.
[197] M. Katoh,et al. Notch signaling in gastrointestinal tract (review). , 2007, International journal of oncology.
[198] Y. Taché,et al. Central nervous system action of calcitonin to alter experimental gastric ulcers in rats. , 1988, Gastroenterology.
[199] Robert C. Wolpert,et al. A Review of the , 1985 .
[200] P Gangadharan,et al. Statistical considerations. , 1973, Indian journal of cancer.