ArticlePDF Available

Abstract

Over the last two decades, remarkable progress has been made in understanding the etiology and pathophysiology of diseases [...]
Journal of
Clinical Medicine
Editorial
Small Bowel—Key Player in Health and Disease
Wojciech Marlicz 1,* and Anastasios Koulaouzidis 2
1Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland
2Centre for Liver & Digestive Disorders, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK;
akoulaouzidis@hotmail.com
*Correspondance: marlicz@hotmail.com
Received: 7 October 2019; Accepted: 14 October 2019; Published: 21 October 2019


Over the last two decades, remarkable progress has been made in understanding the etiology
and pathophysiology of diseases. New discoveries emphasize the importance of the small bowel
(SB) ‘ecosystem’ in the pathogenesis of acute and chronic illness alike. Emerging factors, such as
microbiome, stem and progenitor cells, innate intestinal immunity, and the enteric nervous system,
along with mucosal and endothelial barriers, play a key role in the development of gastrointestinal (GI)
and extra-GI diseases. The results of other studies point also towards a link between the digestive tract
and common non-communicable diseases, such as obesity and cancer. These discoveries unravel novel
dimensions of uncertainty in the area of clinical decision-making motivating researchers to search for
novel diagnostic and therapeutic solutions.
Recent studies unravel the role of the poorly-understood complexity of the SB. Insights into
its critical physiologic and pathophysiologic role in metabolic homeostasis and its potential role as
a driver of obesity, insulin resistance, and subsequent type 2 diabetes mellitus (T2DM) have been
revealed [
1
]. For example, bypassing the proximal small intestine by means of bariatric surgery results
in a significant metabolic benefit to an individual undergoing such a procedure. Moreover, endoscopic
procedures aimed at placing devices separating luminal contents from the duodenal mucosa result in
modest weight loss and improvement in glucose homeostasis [1].
Another endoscopic treatment which aims at resurfacing duodenal mucosa (DMR, duodenal
mucosal resurfacing), leads to improvement in glycemia and insulin resistance in patients with
T2DM [
2
]. It is not surprising when we recall that SB endocrine cells secrete glycemia-regulating
incretin hormones (e.g., glucagon-like peptide, GLP-1), and this process is dependent on food content
in the intestinal lumen. Moreover, bile acids with various targets in the liver and small intestine (e.g.,
farnesoid receptor, FXR) together with a plethora of other small signaling molecules act in concert in
regulating metabolic and digestive GI function.
The human digestive tract and enteric nervous system (ENS) communicate with the central nervous
system (CNS) through the gut-brain axis (GBA). This bidirectional communication involves diverse
neural networks through the X cranial vagal nerve—dorsal roots of the sympathetic/parasympathetic
nervous system. Important roles in the regulation of the gut-brain axis are played by: (i) the
hypothalamus-pituitary-adrenal axis (HPA), (ii) the stress hormones (cortisol), (iii) the short-chain fatty
acids (SCFAs), and (iv) the gut microbiota. The intestinal barrier, another important part of GBA, is
composed of: (i) goblet cells derived mucus, (ii) microbiota, (iii) epithelial cells, (iv) endothelial cells,
(v) lymphatic vessels, and v) enterocytes’ tight cellular junctions. Of interest, the structure and function
of the intestinal barrier resemble that of the blood-brain barrier (BBB). The gut-brain communication is
mediated via blood, portal/hepatic circulation, and the bone marrow [3].
Recently, it has been described that the alterations of the intestinal barrier play a crucial role in the
pathology of several human inflammatory and autoimmune diseases. Mohanan et al. documented
the crucial role of the C1orf106 inflammatory bowel disease (IBD) susceptibility gene in stabilizing
intestinal barrier function and intestinal inflammation [
4
]. Manfredo Vieira et al. evidenced the role of
J. Clin. Med. 2019,8, 1748; doi:10.3390/jcm8101748 www.mdpi.com/journal/jcm
J. Clin. Med. 2019,8, 1748 2 of 5
pathobionts in the process of intestinal barrier alterations, which were followed by their translocation to
lymph nodes and the hepatic portal system, triggering systemic lupus erythematosus (SLE) [
5
]. Thaiss
et al. reported that hyperglycemia leads to disruption of the intestinal barrier followed by intestinal
inflammation and systemic infection [
6
]. Spadoni et al. recently documented that the presence of
the gut-vascular barrier (GVB) in the small intestine controls the dissemination of bacteria into the
bloodstream [
7
]. The authors reported a decrease of the wnt/beta catenin-inducible gene Axin2 (a
marker of stem cell renewal) in gut endothelium under the presence of Salmonella typhimirum in the
SB. The GVB was modified in patients with coeliac disease (CD) with altered serum transaminases,
which suggests that GVB deterioration may be responsible for liver damage in CD patients [
7
]. It has
been shown that disruption of epithelial and vascular barriers in the intestine were early events in
non-alcoholic steatohepatitis (NASH), and GVB leakage marker could be identified in colonic biopsies
in patients with NASH [
8
]. Of importance, SB epithelial and vascular barriers are FXR-controlled, which
opens avenues to clinical trials aimed at investigation of novel FXR-agonists as future therapeutics [
9
].
Of interest, intestinal barriers could be monitored
in vivo
with the aid of confocal laser endomicroscopy
(CLE) [10,11].
Therefore, the SB is considered a key player in metabolic disease development [
12
], including
diabetes mellitus and NASH, and other diet-related disorders such as coeliac and non-coeliac
enteropathies. Another major field is drug metabolism and its interaction with small bowel
microbiome [
13
]. Moreover, the emergence of gut-brain, gut-liver, and gut-blood barriers point
towards the important role of the SB in the pathogenesis of previously unthought and GI-unrelated
conditions such as neurodegenerative and cardiovascular disease [
14
,
15
]. The SB remains an organ
that is dicult to fully access and assess and accurate diagnosis often poses a clinical challenge.
Undoubtedly, the therapeutic potential remains untapped. Therefore, it is now time to direct more
of our interest towards the SB and unravel the interplay between the SB and other GI and non-GI
related diseases.
In this Journal of Clinical Medicine Special Issue, “Diagnosis and Treatment of Small Bowel
Disorders”, several groups of investigators contributed their knowledge to the field of SB research by
presenting original papers and reviews.
Enaud et al. [
16
] describe original observations by utilizing next-generation sequencing (NGS),
that intestinal inflammation in children with Cystic Fibrosis (CF) was associated with alterations of
microbiota similar to those observed in Crohn’s disease (CrDs). Authors for the first time applied novel
CrDs Microbial-Dysbiosis index in CF patients and pointed towards the importance of gut-lung axis
in CF prognosis [
16
]. Of interest, intestinal inflammation was associated with previous intravenous
antibiotic courses for CF [
13
]. This observation is important as global awareness of antibiotic resistance
rises. Nakamura et al. [
17
] sought to evaluate the validity of using capsule endoscopy (CE) to monitor
the eect of medical treatment on SB mucosal healing in post-operative CrDs patients, regardless of the
presence of clinical symptoms. Although the significance of endoscopic monitoring has been widely
accepted in CrDs, to date, only a few studies looked at the validity and eectiveness of escalating
treatment for patients in clinical remission but with endoscopically visible active mucosal lesion.
The authors demonstrated that CrDs patients in clinical remission with ongoing intestinal inflammation
at the time of the CE could benefit from additional treatment. This study motivates physicians to
optimize treatment plans for asymptomatic CD patients.
Several characteristics of SB lesions (e.g., mucosal disruption, bleeding, irregular surface, polypoid
appearance, color, delayed passage, white villi, and invagination) have been described to better predict
SB lesions, such as intestinal bulges, masses, and tumors. The lack of precise features in characterizing
these lesions places a limitation on the accuracy of CE diagnosis. Therefore, Min et al. [
18
] in their
retrospective study, evaluated the utility of an additional morphologic criterion, the mucosal protrusion
angle (MPA), which was defined as the angle between a SB protruding lesion and its surrounding
mucosa. The authors documented that MPA was a simple and useful tool for dierentiating between
J. Clin. Med. 2019,8, 1748 3 of 5
intestinal true masses and non-significant bulges [
18
]. Their observation creates a useful extra tool for
those who are faced with the question of ‘mass or bulge?’
SB microbiota alterations have also been implicated in the pathogenesis of surgical site infections
(SSIs) and surgery-related complications (SRCs). Skonieczna- ˙
Zydecka et al. [
19
] conducted a systematic
review with meta-analysis and meta-regression of randomized clinical trials investigating the ecacy
of probiotics and synbiotics to counteract SSIs and SRCs in patients under various surgical treatments.
The authors aimed to determine the mechanisms behind probiotic/synbiotic action. Their meta-analysis
revealed that probiotics/synbiotics administration prior and at a time of major abdominal surgery, leads
to a reduction in the incidence of SSIs and SRCs (e.g., abdominal distension, diarrhea, pneumonia, sepsis,
urinary tract infection, postoperative pyrexia). Furthermore, probiotics/synbiotics were associated
with shortening of the duration of antibiotic therapy and hospital stay. Based on current evidence, the
action of probiotics/synbiotics in surgical patients seems to be exerted via modulation of gut-immune
response and production of short-chain fatty acids (SCFAs) [19].
Included also in this special issue, Singh et al. [
20
] comprehensively reviewed the pros and cons of
biomarkers in coeliac disease and summarized the current status of coeliac disease screening, diagnosis,
and monitoring. The review could guide clinicians in diagnosis and monitoring of patients with
coeliac disease. As biomarkers allow for smart targeted-screening, similarly imaging spectroscopy (a
combination of digital imaging and spectroscopy, also known as hyperspectral/multispectral (HS/MS)
imaging (HSI/MSI) technology) allows for smart tissue visualization beyond the limitations of the
human eye. HSI has been utilized for various research purposes including: (i) food quality inspection,
(ii) optimization of the recycling process, (iii) art painting renovations, (iv) geology and minerals
inspection, (v) soil evaluation and (vi) plant response to stress. HSI has also been evolving in the field
of medical research in gastroenterology, pathology, and surgery. This modality was used to generate
alternative visualization of tissues, abdominal organ dierentiation, identification of surgical site
resection, and abdominal ischemia to name a few. Ortega et al. [
21
] in their thorough review provided
a detailed summary of the most relevant research work in the field of gastroenterology using HSI.
Last but not least, Skonieczna- ˙
Zydecka et al. [
22
] in a narrative review published in this issue of
Journal of Clinical Medicine, discussed involvements of GBA deregulation in the origin of brain-gut
disorders. The authors hypothesized that stem cell-host microbiome cross-talk was potentially involved
in GBA disorders. Interestingly, patients with inflammatory bowel disease (IBD) have an elevated
risk of mental illness, and depression increases the risk of IBD. Of key interest, are observations
that multiple drugs are known to induce metabolic malfunctions, possibly through alterations of SB
milieu. These alterations result in body weight gain, metabolic disturbances, and suppression of
metabolic resting rate. The authors in their comprehensive review presented the current state of the art
knowledge of the role of GBA in GI and psychiatric comorbidities. The current evidence supports
the notion that an injury to the intestinal mucosa can result in significant, though delayed, metabolic
consequences that may seriously aect the health of an individual. Future investigations [
23
] into the
pathophysiology of host-microbe interactions should focus on the small bowel [
24
,
25
], which is still
relatively inaccessible. Therefore, the research is challenging but necessary to pave the way to new
findings and solutions in the clinical area of the small bowel [26] and beyond [27].
Dr Marlicz and Dr Koulaouzidis are Guest Editors of this Special Issue of the Journal of
Clinical Medicine (ISSN 2077-03830), which belongs to the journal’s section Gastroenterology and
Hepato-Pancreato-Biliary Medicine.
Conflicts of Interest: The authors declare that there is no conflict of interest.
References
1.
Van Baar, A.C.G.; Nieuwdorp, M.; Holleman, F.; Soeters, M.R.; Groen, A.K.; Bergman, J.J.G.H.M.
The Duodenum harbors a Broad Untapped Therapeutic Potential. Gastroenterology
2018
,154, 773–777.
[CrossRef]
J. Clin. Med. 2019,8, 1748 4 of 5
2.
Van Baar, A.C.G.; Holleman, F.; Crenier, L.; Haidry, R.; Magee, C.; Hopkins, D.; Rodriguez Grunert, L.;
Galvao Neto, M.; Vignolo, P.; Hayee, B.; et al. Duodenal mucosal resurfacing for the treatment of type 2
diabetes mellitus: One year results from the first international, open-label, prospective, multicentre study.
Gut 2019. [CrossRef]
3.
Odenwald, M.A.; Turner, J.R. The intestinal epithelial barrier: A therapeutic target? Nat. Rev. Gastroenterol.
Hepatol. 2017,14, 9–21. [CrossRef] [PubMed]
4.
Mohanan, V.; Nakata, T.; Desch, A.N.; L
é
vesque, C.; Boroughs, A.; Guzman, G.; Cao, Z.; Creasey, E.; Yao, J.;
Boucher, G.; et al. C1orf106 is a colitis risk gene that regulates stability of epithelial adherens junctions.
Science 2018,359, 1161–1166. [CrossRef] [PubMed]
5.
Vieira, S.M.; Hiltensperger, M.; Kumar, V.; Zegarra-Ruiz, D.; Dehner, C.; Khan, N.; Costa, F.R.C.; Tiniakou, E.;
Greiling, T.; Ru, W.; et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans.
Science 2018,359, 1156–1161. [CrossRef] [PubMed]
6.
Thaiss, C.A.; Levy, M.; Grosheva, I.; Zheng, D.; Soer, E.; Blacher, E.; Braverman, S.; Tengeler, A.C.; Barak, O.;
Elazar, M.; et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science
2018,359, 1376–1383. [CrossRef]
7.
Spadoni, I.; Zagato, E.; Bertocchi, A.; Paolinelli, R.; Hot, E.; Di Sabatino, A.; Caprioli, F.; Bottiglieri, L.;
Oldani, A.; Viale, G.; et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science
2015,350, 830–834. [CrossRef]
8.
Mouries, J.; Brescia, P.; Silvestri, A.; Spadoni, I.; Sorribas, M.; Wiest, R.; Mileti, E.; Galbiati, M.; Invernizzi, P.;
Adorini, L.; et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic
steatohepatitis development. J. Hepatol. 2019. [CrossRef]
9.
Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.;
Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. NASH Clinical Research Network. Farnesoid X
nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre,
randomised, placebo-controlled trial. Lancet 2015,385, 956–965. [CrossRef]
10.
Sorribas, M.; Jakob, M.O.; Yilmaz, B.; Li, H.; Stutz, D.; Noser, Y.; de Gottardi, A.; Moghadamrad, S.;
Hassan, M.; Albillos, A.; et al. FXR-modulates the gut-vascular barrier by regulating the entry sites for
bacterial translocation in experimental cirrhosis. J. Hepatol. 2019. [CrossRef]
11.
Fritscher-Ravens, A.; Pflaum, T.; Mösinger, M.; Ruchay, Z.; Röcken, C.; Milla, P.J.; Das, M.; Böttner, M.;
Wedel, T.; Schuppan, D. Many Patients with Irritable Bowel Syndrome Have Atypical Food Allergies Not
Associated with Immunoglobulin E. Gastroenterology 2019,157, 109–118. [CrossRef] [PubMed]
12.
Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation.
Nat. Rev. Immunol. 2019. [CrossRef] [PubMed]
13.
Skonieczna- ˙
Zydecka, K.; Łoniewski, I.; Misera, A.; Stachowska, E.; Maciejewska, D.; Marlicz, W.; Galling, B.
Second-generation antipsychotics and metabolism alterations: A systematic review of the role of the gut
microbiome. Psychopharmacology 2019,236, 1491–1512. [CrossRef] [PubMed]
14.
Tanaka, M.; Itoh, H. Hypertension as a Metabolic Disorder and the Novel Role of the Gut. Curr. Hypertens
Rep. 2019,21, 63. [CrossRef] [PubMed]
15.
Kowalski, K.; Mulak, A. Brain-Gut-Microbiota Axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil.
2019
,
25, 48–60. [CrossRef]
16.
Enaud, R.; Hooks, K.B.; Barre, A.; Barnetche, T.; Hubert, C.; Massot, M.; Bazin, T.; Clouzeau, H.; Bui, S.;
Fayon, M.; et al. Intestinal Inflammation in Children with Cystic Fibrosis Is Associated with Crohn’s-Like
Microbiota Disturbances. J. Clin. Med. 2019,8, 645. [CrossRef]
17.
Nakamura, M.; Yamamura, T.; Maeda, K.; Sawada, T.; Mizutani, Y.; Ishikawa, T.; Furukawa, K.; Ohno, E.;
Kawashima, H.; Miyahara, R.; et al. Nagoya University Crohn’s Disease Study Group. Validity of Capsule
Endoscopy in Monitoring Therapeutic Interventions in Patients with Crohn’s Disease. J. Clin. Med.
2018
,
7, 311. [CrossRef]
18.
Min, M.; Noujaim, M.G.; Green, J.; Schlieve, C.R.; Vaze, A.; Cahan, M.A.; Cave, D.R. Role of Mucosal
Protrusion Angle in Discriminating between True and False Masses of the Small Bowel on Video Capsule
Endoscopy. J. Clin. Med. 2019,8, 418. [CrossRef]
J. Clin. Med. 2019,8, 1748 5 of 5
19.
Skonieczna- ˙
Zydecka, K.; Kaczmarczyk, M.; Łoniewski, I.; Lara, L.F.; Koulaouzidis, A.; Misera, A.;
Maciejewska, D.; Marlicz, W. A Systematic Review, Meta-Analysis, and Meta-Regression Evaluating
the Ecacy and Mechanisms of Action of Probiotics and Synbiotics in the Prevention of Surgical Site
Infections and Surgery-Related Complications. J. Clin. Med. 2018,7, 556. [CrossRef]
20.
Singh, A.; Pramanik, A.; Acharya, P.; Makharia, G.K. Non-Invasive Biomarkers for Celiac Disease. J. Clin. Med.
2019,8, 885. [CrossRef]
21.
Ortega, S.; Fabelo, H.; Iakovidis, D.K.; Koulaouzidis, A.; Callico, G.M. Use of Hyperspectral/Multispectral
Imaging in Gastroenterology. Shedding Some Dierent Light into the Dark. J. Clin. Med.
2019
,8, 36.
[CrossRef] [PubMed]
22.
Skonieczna- ˙
Zydecka, K.; Marlicz, W.; Misera, A.; Koulaouzidis, A.; Łoniewski, I. Microbiome—The Missing
Link in the Gut-Brain Axis: Focus on Its Role in Gastrointestinal and Mental Health. J. Clin. Med.
2018
,7, 521.
[CrossRef] [PubMed]
23.
Pełka-Wysiecka, J.; Kaczmarczyk, M.; B ˛aba-Kubi´s, A.; Li´skiewicz, P.; Wro´nski, M.; Skonieczna- ˙
Zydecka, K.;
Marlicz, W.; Misiak, B.; Starzy ´nska, T.; Kucharska-Mazur, J.; et al. Analysis of Gut Microbiota and Their
Metabolic Potential in Patients with Schizophrenia Treated with Olanzapine: Results from a Six-Week
Observational Prospective Cohort Study. J. Clin. Med. 2019,8, 1605. [CrossRef] [PubMed]
24.
Zhong, L.; Shanahan, E.R.; Raj, A.; Koloski, N.A.; Fletcher, L.; Morrison, M.; Walker, M.M.; Talley, N.J.;
Holtmann, G. Dyspepsia and the microbiome: Time to focus on the small intestine. Gut
2017
,66, 1168–1169.
[CrossRef] [PubMed]
25.
Vuik, F.; Dicksved, J.; Lam, S.Y.; Fuhler, G.M.; van der Laan, L.; van de Winkel, A.; Konstantinov, S.R.;
Spaander, M.; Peppelenbosch, M.P.; Engstrand, L.; et al. Composition of the mucosa-associated microbiota
along the entire gastrointestinal tract of human individuals. United Eur. Gastroenterol. J.
2019
,7, 897–907.
[CrossRef] [PubMed]
26.
Quigley, E.M.M. Symptoms and the small intestinal microbiome—The unknown explored. Nat. Rev.
Gastroenterol. Hepatol. 2019,16, 457–458. [CrossRef]
27.
Lynch, S.V.; Ng, S.C.; Shanahan, F.; Tilg, H. Translating the gut microbiome: Ready for the clinic? Nat. Rev.
Gastroenterol. Hepatol. 2019. [CrossRef]
©
2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... In 1915, Walter Alwarez of the Mayo Clinic popularized the term "functional dyspepsia", the most common disorder from the group of functional disorders. Reported dyspeptic ailments are not correlated with specific physiological disorders, although a relationship has been found between the symptoms of premature satiety and disorders of gastric accommodation, in gastroduodenitis, genetic studies are also carried out, or links between bacterial inflammation of the upper and lower gastrointestinal tract and functional disorders [17][18][19][20][21][22][23][24]. There are no objective diagnostic tools; the symptomatic criteria for functional disorders based on the Roman Criteria are expert consensus excluding organic causes of ailments [25][26][27]. ...
Article
Full-text available
Introduction There are many studies on the influence of psychological factors in the appearance of symptoms and their treatment among gastroenterological patients. It is increasingly indicated that these factors are of great importance also for the quality of life of people struggling with a chronic disease. Aim To evaluate personality traits and emotional disorders in female patients with gastrointestinal conditions such as functional dyspepsia (FD), irritable bowel syndrome (IBS), or inflammatory bowel disease (IBD). Material and methods The sample of 28 patients was verified in terms of the disease using the GAST questionnaire and assessed by personality questionnaires and psychological tests: the Spielberger State-Trait Anxiety Inventory (STAI), EAS Temperament Survey, Eysenck Personality Inventory (EPQ-R), Coping Inventory for Stressful Situations (CISS); Beliefs about Pain Control Questionnaire (BPCQ), General Self-efficacy Scale (GSES), and Satisfaction with Life Scale (SWLS). Results The control group was recruited from female university students declaring full health. The conducted statistical analysis showed that there is a significant relationship between personality traits, psychological predispositions, and both the experience of illness and satisfaction with life among this specific group of patients. Conclusions This pilot study demonstrated the need for a personalized approach to gastroenterological patients, also based on their personality characteristics. Such an approach may increase the effectiveness of therapy and bring benefits in long-term treatment.
Chapter
Severe small bowel (SB) dysmotility is often characterized by chronic disabling gastrointestinal (GI) symptoms, associated with dilation of some part of the SB and often inconclusive results of routine endoscopic and radiological investigations or even surgical exploration. Researchers have shown real interest in the last 15 years in the investigation of GI symptoms related to motility abnormalities with wireless capsules. In this chapter, major contributions in this field are introduced and summarized. The main aim is to show first how Computer Vision technics and Artificial Intelligence algorithms can enhance the analysis of the sequences of images extracted from a wireless capsule endoscopy examination for motility analysis, and second, to introduce recent ad hoc capsule technologies making it possible for 3D characterization of the intestinal tract motility.
Article
Full-text available
Accumulating evidence indicates the potential effect of microbiota on the pathogenesis and course of schizophrenia. However, the effects of olanzapine, second-generation antipsychotics, on gut microbiota have not been investigated in humans. This study aimed to analyze fecal microbiota in schizophrenia patients treated with olanzapine during six weeks of their hospital stay. After a seven-day washout from all psychotropic medications, microbiota compositions were evaluated at baseline and after six weeks of hospitalization using 16S rRNA sequencing. The study was conducted in 20 inpatients, who followed the same hospital routine and received 5-20 mg daily doses of olanzapine. Olanzapine treatment was associated with clinical improvements in all patients and significant increases in body mass index in females, but not changes in gut microbiota compositions and predicted function. The severity of symptoms at the beginning of treatment varied in accordance with the predicted metabolic activity of the bacteria. The present findings indicate that the microbiota of schizophrenia patients is highly individual and has different taxonomical (Type 1, with a predominance of Prevotella, and Type 2 with a higher abundance of Bacteroides, Blautia and Clostridium) and functional clusters, and it does not change following six weeks of olanzapine therapy; in addition, the microbiota is not associated with either the weight gain observed in women or the effectiveness of olanzapine therapy.
Article
Full-text available
Background & aims: Fatty liver disease, including non-alcoholic fatty liver (NAFLD) and steatohepatitis (NASH), has been associated with increased intestinal barrier permeability and translocation of bacteria or bacterial products into the blood circulation. In this study, we aimed to unravel the role of both intestinal barrier integrity and microbiota in NAFLD/NASH development. Methods: C57BL/6J mice were fed with high-fat diet (HFD) or methionine-choline-deficient diet for 1 week or longer to recapitulate aspects of NASH (steatosis, inflammation, insulin resistance). Genetic and pharmacological strategies were then used to modulate intestinal barrier integrity. Results: We show that disruption of the intestinal epithelial barrier and gut vascular barrier (GVB) are early events in NASH pathogenesis. Mice fed HFD for only 1 week undergo a diet-induced dysbiosis that drives GVB damage and bacterial translocation into the liver. Fecal microbiota transplantation from HFD-fed mice into specific pathogen-free recipients induces GVB damage and epididymal adipose tissue enlargement. GVB disruption depends on interference with the WNT/β-catenin signaling pathway, as shown by genetic intervention driving β-catenin activation only in endothelial cells, preventing GVB disruption and NASH development. The bile acid analogue and farnesoid X receptor agonist obeticholic acid (OCA) drives β-catenin activation in endothelial cells. Accordingly, pharmacologic intervention with OCA protects against GVB disruption, both as a preventive and therapeutic agent. Importantly, we found upregulation of the GVB leakage marker in the colon of patients with NASH. Conclusions: We have identified a new player in NASH development, the GVB, whose damage leads to bacteria or bacterial product translocation into the blood circulation. Treatment aimed at restoring β-catenin activation in endothelial cells, such as administration of OCA, protects against GVB damage and NASH development. Lay summary: The incidence of fatty liver disease is reaching epidemic levels in the USA, with more than 30% of adults having NAFLD (non-alcoholic fatty liver disease), which can progress to more severe non-alcoholic steatohepatitis (NASH). Herein, we show that disruption of the intestinal epithelial barrier and gut vascular barrier are early events in the development of NASH. We show that the drug obeticholic acid protects against barrier disruption and thereby prevents the development of NASH, providing further evidence for its use in the prevention or treatment of NASH.
Article
Full-text available
Background The duodenum has become a metabolic treatment target through bariatric surgery learnings and the specific observation that bypassing, excluding or altering duodenal nutrient exposure elicits favourable metabolic changes. Duodenal mucosal resurfacing (DMR) is a novel endoscopic procedure that has been shown to improve glycaemic control in people with type 2 diabetes mellitus (T2D) irrespective of body mass index (BMI) changes. DMR involves catheter-based circumferential mucosal lifting followed by hydrothermal ablation of duodenal mucosa. This multicentre study evaluates safety and feasibility of DMR and its effect on glycaemia at 24 weeks and 12 months. Methods International multicentre, open-label study. Patients (BMI 24–40) with T2D (HbA1c 59–86 mmol/mol (7.5%–10.0%)) on stable oral glucose-lowering medication underwent DMR. Glucose-lowering medication was kept stable for at least 24 weeks post DMR. During follow-up, HbA1c, fasting plasma glucose (FPG), weight, hepatic transaminases, Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), adverse events (AEs) and treatment satisfaction were determined and analysed using repeated measures analysis of variance with Bonferroni correction. Results Forty-six patients were included of whom 37 (80%) underwent complete DMR and 36 were finally analysed; in remaining patients, mainly technical issues were observed. Twenty-four patients had at least one AE (52%) related to DMR. Of these, 81% were mild. One SAE and no unanticipated AEs were reported. Twenty-four weeks post DMR (n=36), HbA1c (−10±2 mmol/mol (−0.9%±0.2%), p<0.001), FPG (−1.7±0.5 mmol/L, p<0.001) and HOMA-IR improved (−2.9±1.1, p<0.001), weight was modestly reduced (−2.5±0.6 kg, p<0.001) and hepatic transaminase levels decreased. Effects were sustained at 12 months. Change in HbA1c did not correlate with modest weight loss. Diabetes treatment satisfaction scores improved significantly. Conclusions In this multicentre study, DMR was found to be a feasible and safe endoscopic procedure that elicited durable glycaemic improvement in suboptimally controlled T2D patients using oral glucose-lowering medication irrespective of weight loss. Effects on the liver are examined further. Trial registration number NCT02413567
Article
Full-text available
Background & aims: Pathological bacterial translocation (PBT) in cirrhosis is the hallmark of spontaneous bacterial infections, increasing mortality several-fold. Increased intestinal permeability is known to contribute to PBT in cirrhosis, although the role of the mucus layer has not been addressed in detail. A clear route of translocation for luminal intestinal bacteria is yet to be defined, but we hypothesize that the recently described gut-vascular barrier (GVB) is impaired in experimental portal hypertension, leading to increased accessibility of the vascular compartment for translocating bacteria. Materials: Cirrhosis was induced in mouse models using bile-duct ligation (BDL) and CCl4. Pre-hepatic portal-hypertension was induced by partial portal vein ligation (PPVL). Intestinal permeability was compared in these mice after GFP-Escherichia coli or different sized FITC-dextrans were injected into the intestine. Results: Healthy and pre-hepatic portal-hypertensive (PPVL) mice lack translocation of FITC-dextran and GFP-E. coli from the small intestine to the liver, whereas BDL and CCl4-induced cirrhotic mice demonstrate pathological translocation, which is not altered by prior thoracic-duct ligation. The mucus layer is reduced in thickness, with loss of goblet cells and Muc2-staining and expression in cirrhotic but not PPVL mice. These changes are associated with bacterial overgrowth in the inner mucus layer and pathological translocation of GFP-E. coli through the ileal epithelium. GVB is profoundly altered in BDL and CCl4-mice with Ileal extravasation of large-sized 150 kDa-FITC-dextran, but only slightly altered in PPVL mice. This pathological endothelial permeability and accessibility in cirrhotic mice is associated with augmented expression of PV1 in intestinal vessels. OCA but not fexaramine stabilizes the GVB, whereas both FXR-agonists ameliorate gut to liver translocation of GFP-E. coli. Conclusions: Cirrhosis, but not portal hypertension per se, grossly impairs the endothelial and muco-epithelial barriers, promoting PBT to the portal-venous circulation. Both barriers appear to be FXR-modulated, with FXR-agonists reducing PBT via the portal-venous route. Lay summary: For intestinal bacteria to enter the systemic circulation, they must cross the mucus and epithelial layer, as well as the gut-vascular barrier. Cirrhosis disrupts all 3 of these barriers, giving bacteria access to the portal-venous circulation and thus, the gut-liver axis. Diminished luminal bile acid availability, cirrhosis and the associated reduction in farnesoid x receptor (FXR) signaling seem, at least partly, to mediate these changes, as FXR-agonists reduce bacterial translocation via the portal-venous route to the liver in cirrhosis.
Article
Full-text available
Purpose of Review Hypertension is related to impaired metabolic homeostasis and can be regarded as a metabolic disorder. This review presents possible mechanisms by which metabolic disorders increase blood pressure (BP) and discusses the importance of the gut as a novel modulator of BP. Recent Findings Obesity and high salt intake are major risk factors for hypertension. There is a hypothesis of “salt-induced obesity”; i.e., high salt intake may tie to obesity. Heightened sympathetic nervous system (SNS) activity, especially in the kidney and brain, increases BP in obese patients. Adipokines, including adiponectin and leptin, and renin-angiotensin-aldosterone system (RAAS) contribute to hypertension. Adiponectin induced by a high-salt diet may decrease sodium/glucose cotransporter (SGLT) 2 expression in the kidney, which results in reducing BP. High salt can change secretions of adipokines and RAAS-related components. Evidence has been accumulating linking the gastrointestinal tract to BP. Glucagon-like peptide-1 (GLP-1) and ghrelin decrease BP in both rodents and humans. The sweet taste receptor in enteroendocrine cells increases SGLT1 expression and stimulates sodium/glucose absorption. Roux-en-Y gastric bypass improves glycemic and BP control due to reducing the activity of SGLT1. Na/H exchanger isoform 3 (NHE3) increases BP by stimulating the intestinal absorption of sodium. Gastrin functions as an intestinal sodium taste sensor and inhibits NHE3 activity. Intestinal mineralocorticoid receptors also regulate sodium absorption and BP due to changing ENaC activity. Gastric sensing of sodium induces natriuresis, and gastric distension increases BP. Changes in the composition and function of gut microbiota contribute to hypertension. A high-salt/fat diet may disrupt the gut barrier, which results in systemic inflammation, insulin resistance, and increased BP. Gut microbiota regulates BP by secreting vasoactive hormones and short-chain fatty acids. BP-lowering effects of probiotics and antibiotics have been reported. Bariatric surgery improves metabolic disorders and hypertension due to increasing GLP-1 secretion, decreasing leptin secretion and SNS activity, and changing gut microbiome composition. Strategies targeting the gastrointestinal system may be therapeutic options for improving metabolic abnormalities and reducing BP in humans. Summary SNS, brain, adipocytes, RAAS, the kidney, the gastrointestinal tract, and microbiota play important roles in regulating BP. Most notably, the gut could be a novel target for treatment of hypertension as a metabolic disorder.
Article
Full-text available
Once thought to be uncommon, celiac disease has now become a common disease globally. While avoidance of the gluten-containing diet is the only effective treatment so far, many new targets are being explored for the development of new drugs for its treatment. The endpoints of therapy include not only reversal of symptoms, normalization of immunological abnormalities and healing of mucosa, but also maintenance of remission of the disease by strict adherence of the gluten-free diet (GFD). There is no single gold standard test for the diagnosis of celiac disease and the diagnosis is based on the presence of a combination of characteristics including the presence of a celiac-specific antibody (anti-tissue transglutaminase antibody, anti-endomysial antibody or anti-deamidated gliadin peptide antibody) and demonstration of villous abnormalities. While the demonstration of enteropathy is an important criterion for a definite diagnosis of celiac disease, it requires endoscopic examination which is perceived as an invasive procedure. The capability of prediction of enteropathy by the presence of the high titer of anti-tissue transglutaminase antibody led to an option of making a diagnosis even without obtaining mucosal biopsies. While present day diagnostic tests are great, they, however, have certain limitations. Therefore, there is a need for biomarkers for screening of patients, prediction of enteropathy, and monitoring of patients for adherence of the gluten-free diet. Efforts are now being made to explore various biomarkers which reflect different changes that occur in the intestinal mucosa using modern day tools including transcriptomics, proteomics, and metabolomics. In the present review, we have discussed comprehensively the pros and cons of available biomarkers and also summarized the current status of emerging biomarkers for the screening, diagnosis, and monitoring of celiac disease.
Article
Full-text available
Background: Homeostasis of the gastrointestinal tract depends on a healthy bacterial microbiota, with alterations in microbiota composition suggested to contribute to diseases. To unravel bacterial contribution to disease pathology, a thorough understanding of the microbiota of the complete gastrointestinal tract is essential. To date, most microbial analyses have either focused on faecal samples, or on the microbial constitution of one gastrointestinal location instead of different locations within one individual. Objective: We aimed to analyse the mucosal microbiome along the entire gastrointestinal tract within the same individuals. Methods: Mucosal biopsies were taken from nine different sites in 14 individuals undergoing antegrade and subsequent retrograde double-balloon enteroscopy. The bacterial composition was characterised using 16 S rRNA sequencing with Illumina Miseq. Results: At double-balloon enteroscopy, one individual had a caecal adenocarcinoma and one individual had Peutz-Jeghers polyps. The composition of the microbiota distinctively changed along the gastrointestinal tract with larger bacterial load, diversity and abundance of Firmicutes and Bacteroidetes in the lower gastrointestinal tract than the upper gastrointestinal tract, which was predominated by Proteobacteria and Firmicutes. Conclusions: We show that gastrointestinal location is a larger determinant of mucosal microbial diversity than inter-person differences. These data provide a baseline for further studies investigating gastrointestinal microbiota-related disease.
Article
Research into the gut microbiota and its role in health and disease has expanded rapidly in the past two decades. However, much of the early focus has been on cataloguing the microorganisms present, identifying correlations between microbial species and disease and using preclinical animal models to understand phenotypes. Now efforts are under way to provide functional insights into the gut microbiota and its mechanisms of action, improve understanding of the role of the microbiota beyond the gut and advance the development of microbiota-based therapeutics so that the microbiome can be harnessed in the clinic. In this Viewpoint article, we asked a selection of scientists and clinicians working in the gut microbiome field for their opinions on the major advances in and the challenges and solutions for translating gut microbiome research to the clinic, and where they expect progress to be made in the future.
Article
Low-grade inflammation is the hallmark of metabolic disorders such as obesity, type 2 diabetes and nonalcoholic fatty liver disease. Emerging evidence indicates that these disorders are characterized by alterations in the intestinal microbiota composition and its metabolites, which translocate from the gut across a disrupted intestinal barrier to affect various metabolic organs, such as the liver and adipose tissue, thereby contributing to metabolic inflammation. Here, we discuss some of the recently identified mechanisms that showcase the role of the intestinal microbiota and barrier dysfunction in metabolic inflammation. We propose a concept by which the gut microbiota fuels metabolic inflammation and dysregulation. Here, the authors describe how metabolic disorders, such as type 2 diabetes and nonalcoholic fatty liver disease, are driven by alterations in the composition of the intestinal microbiota and its metabolites, which translocate from the gut across a disrupted intestinal barrier and contribute to metabolic inflammation.
Article
Small intestinal bacterial overgrowth (SIBO) has been linked to common gastrointestinal symptoms; yet its precise definition remains elusive. A new study shows that, when it comes to symptom prediction in functional gastrointestinal disorders, high-throughput sequencing analysis of the microbiome and not jejunal aspiration and culture is the clear winner.