1
|
Liu X, Liu X, Huang Y, Lin C, Chen X, Li Y, Huang Y, Wang Y. Association between dietary index for gut microbiota and diarrhea among US adults: a cross-sectional analysis of NHANES 2005-2010. Front Nutr 2025; 12:1566314. [PMID: 40290658 PMCID: PMC12021626 DOI: 10.3389/fnut.2025.1566314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives Growing attention has been paid to the relationship between the gut microbiota and diarrhea. A recently proposed Dietary Index for Gut Microbiota (DI-GM) reflects the overall dietary quality as it pertains to gut microbiota diversity. However, evidence regarding the association between DI-GM and diarrhea is still lacking. This study aims to investigate the association between DI-GM and the risk of diarrhea. Methods A total of 15,590 adults (≥20 years old) from National Health and Nutrition Examination Survey (NHANES) 2005-2010 were included in this analysis. Diarrhea was defined by self-reported common Bristol Stool Form Scale (BSFS) type 6 or 7, or more than three bowel movements per day. DI-GM comprises 14 food/nutrient components known to be associated with gut microbiota. A higher score indicates a more favorable diet for the gut microbiota. Multivariable logistic regression was used to evaluate the association between DI-GM and diarrhea, with subgroup and sensitivity analyses performed to confirm robustness. Results After adjusting for age, gender, race, physical activity, chronic diseases, and other confounders, each 1-point increase in DI-GM was associated with a 5% reduction in diarrhea risk (adjusted OR = 0.95, 95% CI: 0.91-0.98, p = 0.005). Compared with those who had a DI-GM score of 0-3, individuals with a DI-GM score ≥ 6 demonstrated a significantly lower risk of diarrhea (OR = 0.77, 95% CI: 0.65-0.91, p = 0.002). Subgroup and sensitivity analyses further supported this negative association. Notably, the "beneficial component" was found to have a more pronounced effect on reducing diarrhea risk. Conclusion Based on a large representative population, our findings suggest that a higher DI-GM score is significantly associated with a lower risk of diarrhea, underscoring the importance of overall dietary patterns in maintaining gut function and homeostasis.
Collapse
Affiliation(s)
- Xiaoqiang Liu
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Xiaobo Liu
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Yingxuan Huang
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Chanchan Lin
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Xinqi Chen
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Yingyi Li
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Yisen Huang
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| | - Yubin Wang
- Department of Gastroenterology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
2
|
Qin L, Lv W. Dietary content and eating behavior in ulcerative colitis: a narrative review and future perspective. Nutr J 2025; 24:12. [PMID: 39849464 PMCID: PMC11755847 DOI: 10.1186/s12937-025-01075-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025] Open
Abstract
Ulcerative colitis (UC) has experienced a steady increase in global incidence and prevalence recently. Current research into UC pathogenesis focuses on the complex interplay of genetic and environmental factors with the immune system and gut microbiome, leading to disruption of the intestinal barrier. Normally, the microbiome, intestinal epithelium, and immune system interact to maintain intestinal homeostasis. However, when this equilibrium is disturbed, a harmful cycle of dysbiosis, immune dysregulation, and inflammation emerges, resulting in intestinal barrier dysfunction and UC progression. Among various risk factors, diet significantly influences epithelial barrier integrity and architectural stability through both direct and indirect mechanisms, shaping the entire UC continuum from pre-clinical prevention to active phase treatment and remission maintenance. This review provides insights into the impact of dietary content and eating behaviors on UC, focusing on specific food, food groups, nutrients, and intermittent fasting, while providing a detailed explanation of why the gut microbiota may mediate the sustained effects of diet across all stages of UC. Additionally, it addresses the limitations of current studies, explores underexamined areas in UC dietary research and proposes potential directions for future research and expansion.
Collapse
Affiliation(s)
- Lingxi Qin
- Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Wenliang Lv
- Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Deleu S, Becherucci G, Godny L, Mentella MC, Petito V, Scaldaferri F. The Key Nutrients in the Mediterranean Diet and Their Effects in Inflammatory Bowel Disease: A Narrative Review. Nutrients 2024; 16:4201. [PMID: 39683595 DOI: 10.3390/nu16234201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The gut microbiome, a collection of gut microorganisms, is crucial in the development and progression of inflammatory bowel diseases (IBD). Therefore, diet and dietary interventions are promising strategies to shape the gut microbiota for IBD management. Of all the diets studied in the IBD field, the Mediterranean diet has the least restrictive nature, promoting long-term adherence. The Mediterranean diet is rich in plants, with a high daily intake of fruits and vegetables (high in fiber, antioxidants, and vitamins), olive oil, whole grains, legumes, and nuts. It includes the moderate consumption of animal products such as oily fish (rich in mono- and polyunsaturated fatty acids), dairy products, and poultry, with a limited intake of red meat and processed foods. This diet is associated with a decreased risk of chronic diseases, including IBD. However, the mechanisms of specific nutrients behind these effects in the Mediterranean diet remain under investigation. Therefore, in this review, we aim to provide an overview of the nutrients that are abundant in the Mediterranean diet and their effects on IBD, with a main focus on preclinical evidence. While several nutrients like fructo-oligosaccharide, chitosan, plant-derived protein, polyphenols, omega-3 polyunsaturated fatty acids, and resveratrol have shown potential beneficial effects in preclinical models, clinical evidence is often limited. However, understanding the complex interactions between specific nutrients and IBD is essential to developing a tailored, multidisciplinary, and personalized approach for disease management; therefore, further research is required.
Collapse
Affiliation(s)
- Sara Deleu
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Guia Becherucci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lihi Godny
- Division of Gastroenterology and Nutrition Unit, Rabin Medical Center, Petah-Tikva 49100, Israel
| | - Maria Chiara Mentella
- UOC di Nutrizione Clinica, Dipartimento Scienze Mediche e Chirurgiche Addominali ed Endocrino-Metaboliche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Valentina Petito
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
4
|
Liu T, Ji H, Li Z, Luan Y, Zhu C, Li D, Gao Y, Yan Z. Gut microbiota causally impacts adrenal function: a two-sample mendelian randomization study. Sci Rep 2024; 14:23338. [PMID: 39375408 PMCID: PMC11458771 DOI: 10.1038/s41598-024-73420-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Some studies have reported that the gut microbiota can influence adrenal-related hormone levels. However, the causal effects of the gut microbiota on adrenal function remain unknown. Therefore, we employed a two-sample Mendelian randomization (MR) study to systematically investigate the impact of gut microbiota on the function of different regions of the adrenal gland. The summary statistics for gut microbiota and adrenal-related hormones used in the two-sample MR analysis were derived from publicly available genome-wide association studies (GWAS). In the MR analysis, inverse variance weighting (IVW) was used as the primary method, with MR-Egger, weighted median, and cML-MA serving as supplementary methods for causal inference. Sensitivity analyses such as the MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were used to assess pleiotropy and heterogeneity. We identified 27 causal relationships between 23 gut microbiota and adrenal function using the IVW method. Among these, Sellimonas enhanced the function of the adrenal cortex reticularis zone (beta = 0.008, 95% CI: 0.002-0.013, P = 0.0057). The cML-MA method supported our estimate (beta = 0.009, 95% CI: 0.004-0.013, P = 2 × 10- 4). Parasutterella, Sutterella, and Anaerofilum affect the functioning of different regions of the adrenal gland. Notably, pleiotropy was not observed. Our findings revealed that the gut microbiota is causally associated with adrenal function. This enhances our understanding of the gut-microbiota-brain axis and provides assistance in the early diagnosis and treatment of adrenal-related diseases in clinical practice.
Collapse
Affiliation(s)
- Tonghu Liu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Hongfei Ji
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhiyuan Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yongkun Luan
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Congcong Zhu
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Dongxiao Li
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| | - Yukui Gao
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, Anhui, People's Republic of China.
| | - Zechen Yan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
5
|
Sztolsztener K, Harasim-Symbor E, Chabowski A, Konstantynowicz-Nowicka K. Cannabigerol as an anti-inflammatory agent altering the level of arachidonic acid derivatives in the colon tissue of rats subjected to a high-fat high-sucrose diet. Biomed Pharmacother 2024; 178:117286. [PMID: 39128189 DOI: 10.1016/j.biopha.2024.117286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
Fat and sugar overconsumption is the cause of increasing worldwide incidence of gastrointestinal tract in inflammatory conditions. The intestinal pre-inflammatory alterations are partially reversible, simultaneously inhibiting the predisposition to colitis. Searching for an effective pharmacotherapy for treating inflammatory conditions in the intestine is essential. This study aimed to investigate the effect of cannabigerol (CBG) on the inflammation state in the colon tissue of rats subjected to high-caloric diet. The experiment was conducted on male Wistar rats subjected to a standard or a high-fat high-sucrose diets for six weeks. For the last 14 days, half of rats from both groups received intragastrically cannabigerol solution (30 mg/kg of body mass). The ratio of n-6/n-3 PUFA, the activity of n-6 and n-3 PUFA, and arachidonic acid (AA) content in selected lipid fractions were determined by gas-liquid chromatography. Immunoblotting examined the expression of proteins involved in inflammation development. ELISA kits measured the content of arachidonic acid derivatives. CBG treatment reduced the n-6/n-3 PUFA ratio in TAG fraction and increased the n-3 PUFA pathway activity in almost all lipid fractions. Cannabigerol supplementation decreased AA concentration in PL and TAG. CBG also caused diminishments in the expression of cPLA2, COX-1, COX-2, and 12/15-LOX, which was indirectly correlated with a decreased LTB4 level and an increased LXA4 level. We concluded that cannabigerol has a protective influence on the development of inflammation in the colon tissue under lipid and sugar overload condition, thereby favoring cancer initiation and progression.
Collapse
Affiliation(s)
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland.
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland.
| | | |
Collapse
|
6
|
Sayegh LN, Haddad F, Bou Jaoude L, Fakhoury-Sayegh N, Heraoui GNHA, Nasrallah Z, Chidiac C, Nawfal R, Francis FF, Mourad FH, Hashash JG. Nutritional Profile, Disease Severity, and Quality of Life of Patients with Inflammatory Bowel Disease: A Case-Control Study. Nutrients 2024; 16:1826. [PMID: 38931181 PMCID: PMC11206244 DOI: 10.3390/nu16121826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Diet is thought to play an important role in the clinical course and quality of life (QOL) of patients with inflammatory bowel disease (IBD). However, dietary habits of patients with IBD are still unknown. This case-control study aims to compare the dietary habits of patients with IBD to healthy controls and evaluate differences in disease severity and QOL. MATERIALS AND METHODS Food frequency, severity scores using the Harvey-Bradshaw and Ulcerative colitis activity index, and QOL were assessed using online questionnaires. Dietary habits were compared for patients with active disease and remission and for those with low QOL (LQOL) and high QOL (HQOL). RESULTS We recruited 61 patients with IBD and 101 controls. Significance was set at p = 0.05. Controls consumed significantly more daily calories (2546 vs. 1641, p = 0.001). However, patients with IBD consumed a higher percentage of carbohydrates (50% vs. 45%, p = 0.001), more red meat (p = 0.024), and less fiber, sucrose, and lactose (p = 0.001, 0.001, and 0.036). Patients with active disease had higher lipid intake, lower protein intake, and lower QOL (47 vs. 58, p = 0.001). Dietary differences between LQOL and HQOL mirrored those between active disease and remission. CONCLUSION This study is the first to provide valuable insights into the nutritional profile of Lebanese patients with IBD.
Collapse
Affiliation(s)
- Lea N. Sayegh
- Department of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St. SW, Rochester, MN 55905, USA;
- School of Medicine, American University of Beirut, Bliss St., Beirut P.O. Box 11-0236, Lebanon; (F.H.); (L.B.J.); (Z.N.); (C.C.)
| | - Firas Haddad
- School of Medicine, American University of Beirut, Bliss St., Beirut P.O. Box 11-0236, Lebanon; (F.H.); (L.B.J.); (Z.N.); (C.C.)
| | - Layane Bou Jaoude
- School of Medicine, American University of Beirut, Bliss St., Beirut P.O. Box 11-0236, Lebanon; (F.H.); (L.B.J.); (Z.N.); (C.C.)
| | - Nicole Fakhoury-Sayegh
- Department of Nutrition, Saint Joseph University, Damascus St., Beirut P.O. Box 17-5208, Lebanon; (N.F.-S.); (G.N.H.A.H.)
| | - Gessica N. H. A. Heraoui
- Department of Nutrition, Saint Joseph University, Damascus St., Beirut P.O. Box 17-5208, Lebanon; (N.F.-S.); (G.N.H.A.H.)
| | - Zainab Nasrallah
- School of Medicine, American University of Beirut, Bliss St., Beirut P.O. Box 11-0236, Lebanon; (F.H.); (L.B.J.); (Z.N.); (C.C.)
- Department of Internal Medicine, Indiana University, 1120 W Michigan St., Indianapolis, IN 46202, USA
| | - Charbel Chidiac
- School of Medicine, American University of Beirut, Bliss St., Beirut P.O. Box 11-0236, Lebanon; (F.H.); (L.B.J.); (Z.N.); (C.C.)
- Department of Surgery, Johns Hopkins School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA
| | - Rashad Nawfal
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Bliss Street, Beirut P.O. Box 11-0236, Lebanon;
- Department of Medical Oncology, Dana Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02215, USA
| | - Fadi F. Francis
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh, 200 Lothrop St., Pittsburgh, PA 15261, USA;
| | - Fadi H. Mourad
- Department of Gastroenterology and Hepatology, American University of Beirut Medical Center, Cairo Street, Beirut P.O. Box 11-0236, Lebanon;
| | - Jana G. Hashash
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
7
|
Xu D, Peng Z, Li Y, Hou Q, Peng Y, Liu X. Progress and Clinical Applications of Crohn's Disease Exclusion Diet in Crohn's Disease. Gut Liver 2024; 18:404-413. [PMID: 37842728 PMCID: PMC11096903 DOI: 10.5009/gnl230093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 10/17/2023] Open
Abstract
Crohn's disease is a chronic intestinal inflammatory disorder of unknown etiology. Although the pharmacotherapies for Crohn's disease are constantly updating, nutritional support and adjuvant therapies have recently gained more attention. Due to advancements in clinical nutrition, various clinical nutritional therapies are used to treat Crohn's disease. Doctors treating inflammatory bowel disease can now offer several diets with more flexibility than ever. The Crohn's disease exclusion diet is a widely used diet for patients with active Crohn's disease. The Crohn's disease exclusion diet requires both exclusion and inclusion. Periodic exclusion of harmful foods and inclusion of wholesome foods gradually improves a patient's nutritional status. This article reviews the Crohn's disease exclusion diet, including its structure, mechanisms, research findings, and clinical applications.
Collapse
Affiliation(s)
- Duo Xu
- Departments of Gastroenterology, Xiangya Hospital of Central South University, Changsha, China
| | - Ziheng Peng
- Departments of Gastroenterology, Xiangya Hospital of Central South University, Changsha, China
| | - Yong Li
- Departments of Gastroenterology, Xiangya Hospital of Central South University, Changsha, China
| | - Qian Hou
- Departments of Clinical Nutrition, Xiangya Hospital of Central South University, Changsha, China
| | - Yu Peng
- Departments of Gastroenterology, Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, China
| | - Xiaowei Liu
- Departments of Gastroenterology, Xiangya Hospital of Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| |
Collapse
|
8
|
Zhou GQ, Huang MJ, Yu X, Zhang NN, Tao S, Zhang M. Early life adverse exposures in irritable bowel syndrome: new insights and opportunities. Front Pediatr 2023; 11:1241801. [PMID: 37732013 PMCID: PMC10507713 DOI: 10.3389/fped.2023.1241801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder worldwide. Extensive research has identified multiple factors contributing to its development, including genetic predisposition, chronic infection, gut dysbiosis, aberrant serotonin metabolism, and brain dysfunction. Recent studies have emphasized the critical role of the early life stage as a susceptibility window for IBS. Current evidence suggests that diet can heighten the risk of IBS in offspring by influencing the microbiota composition, intestinal epithelium structure, gene expression, and brain-gut axis. The use of antibiotics during pregnancy and the neonatal period disrupts the normal gut microbiota structure, aligning it with the characteristics observed in IBS patients. Additionally, early life stress impacts susceptibility to IBS by modulating TLR4, NK1, and the hypothalamic-pituitary-adrenal (HPA) axis while compromising the offspring's immune system. Formula feeding facilitates the colonization of pathogenic bacteria in the intestines, concurrently reducing the presence of probiotics. This disruption of the Th1 and Th2 cell balance in the immune system weakens the intestinal epithelial barrier. Furthermore, studies suggest that delivery mode influences the occurrence of IBS by altering the composition of gut microbes. This review aims to provide a comprehensive summary of the existing evidence regarding the impact of adverse early life exposures on IBS during pregnancy, intrapartum, and neonatal period. By consolidating this knowledge, the review enhances our understanding of the direct and indirect mechanisms underlying early life-related IBS and offers new insights and research directions from childhood to adulthood.
Collapse
Affiliation(s)
| | | | | | | | | | - Ming Zhang
- Department of General Practice, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| |
Collapse
|
9
|
Dwiyanto J, Huët MAL, Hussain MH, Su TT, Tan JBL, Toh KY, Lee JWJ, Rahman S, Chong CW. Social demographics determinants for resistome and microbiome variation of a multiethnic community in Southern Malaysia. NPJ Biofilms Microbiomes 2023; 9:55. [PMID: 37573460 PMCID: PMC10423249 DOI: 10.1038/s41522-023-00425-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 08/03/2023] [Indexed: 08/14/2023] Open
Abstract
The prevalence of antibiotic-resistant bacteria in Southeast Asia is a significant concern, yet there is limited research on the gut resistome and its correlation with lifestyle and environmental factors in the region. This study aimed to profile the gut resistome of 200 individuals in Malaysia using shotgun metagenomic sequencing and investigate its association with questionnaire data comprising demographic and lifestyle variables. A total of 1038 antibiotic resistance genes from 26 classes were detected with a mean carriage rate of 1.74 ± 1.18 gene copies per cell per person. Correlation analysis identified 14 environmental factors, including hygiene habits, health parameters, and intestinal colonization, that were significantly associated with the resistome (adjusted multivariate PERMANOVA, p < 0.05). Notably, individuals with positive yeast cultures exhibited a reduced copy number of 15 antibiotic resistance genes. Network analysis highlighted Escherichia coli as a major resistome network hub, with a positive correlation to 36 antibiotic-resistance genes. Our findings suggest that E. coli may play a pivotal role in shaping the resistome dynamics in Segamat, Malaysia, and its abundance is strongly associated with the community's health and lifestyle habits. Furthermore, the presence of yeast appears to be associated with the suppression of antibiotic-resistance genes.
Collapse
Affiliation(s)
- J Dwiyanto
- AMILI, Singapore, 118261, Singapore.
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - M A L Huët
- Faculty of Science, University of Mauritius, Reduit, 80837, Mauritius
| | - M H Hussain
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Malaysia
| | - T T Su
- South East Asia Community Observatory, Segamat, 85000, Malaysia
| | - J B L Tan
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Malaysia
| | - K Y Toh
- AMILI, Singapore, 118261, Singapore
| | - J W J Lee
- AMILI, Singapore, 118261, Singapore
- Department of Medicine, National University Hospital, Singapore, 119228, Singapore
| | - S Rahman
- School of Science, Monash University Malaysia, Bandar Sunway, 47500, Malaysia
- Tropical Medicine and Biology Platform, Monash University Malaysia, Bandar Sunway, 47500, Malaysia
| | - C W Chong
- AMILI, Singapore, 118261, Singapore.
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500, Malaysia.
| |
Collapse
|
10
|
Mitchell SB, Hung YH, Thorn TL, Zou J, Baser F, Gulec S, Cheung C, Aydemir TB. Sucrose-induced hyperglycemia dysregulates intestinal zinc metabolism and integrity: risk factors for chronic diseases. Front Nutr 2023; 10:1220533. [PMID: 37637953 PMCID: PMC10450956 DOI: 10.3389/fnut.2023.1220533] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Objective Zinc is an essential micronutrient that is critical for many physiological processes, including glucose metabolism, regulation of inflammation, and intestinal barrier function. Further, zinc dysregulation is associated with an increased risk of chronic inflammatory diseases such as type II diabetes, obesity, and inflammatory bowel disease. However, whether altered zinc status is a symptom or cause of disease onset remains unclear. Common symptoms of these three chronic diseases include the onset of increased intestinal permeability and zinc dyshomeostasis. The specific focus of this work is to investigate how dietary sources of intestinal permeability, such as high sucrose consumption, impact transporter-mediated zinc homeostasis and subsequent zinc-dependent physiology contributing to disease development. Method We used in vivo subchronic sucrose treatment, ex vivo intestinal organoid culture, and in vitro cell systems. We analyze the alterations in zinc metabolism and intestinal permeability and metabolic outcomes. Results We found that subchronic sucrose treatment resulted in systemic changes in steady-state zinc distribution and increased 65Zn transport (blood-to-intestine) along with greater ZIP14 expression at the basolateral membrane of the intestine. Further, sucrose treatment enhanced cell survival of intestinal epithelial cells, activation of the EGFR-AKT-STAT3 pathway, and intestinal permeability. Conclusion Our work suggests that subchronic high sucrose consumption alters systemic and intestinal zinc homeostasis linking diet-induced changes in zinc homeostasis to the intestinal permeability and onset of precursors for chronic disease.
Collapse
Affiliation(s)
| | - Yu-Han Hung
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Trista Lee Thorn
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Jiaqi Zou
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Filiz Baser
- Molecular Nutrition and Human Physiology Laboratory, Department of Food Engineering, İzmir Institute of Technology, İzmir, Türkiye
| | - Sukru Gulec
- Molecular Nutrition and Human Physiology Laboratory, Department of Food Engineering, İzmir Institute of Technology, İzmir, Türkiye
| | - Celeste Cheung
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | | |
Collapse
|
11
|
Wang YC, Cao Y, Pan C, Zhou Z, Yang L, Lusis AJ. Intestinal cell type-specific communication networks underlie homeostasis and response to Western diet. J Exp Med 2023; 220:213924. [PMID: 36880999 PMCID: PMC10038833 DOI: 10.1084/jem.20221437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/14/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
The small intestine plays a key role in immunity and mediates inflammatory responses to high fat diets. We have used single-cell RNA-sequencing (scRNA-seq) and statistical modeling to examine gaps in our understanding of the dynamic properties of intestinal cells and underlying cellular mechanisms. Our scRNA-seq and flow cytometry studies of different layers of intestinal cells revealed new cell subsets and modeled developmental trajectories of intestinal intraepithelial lymphocytes, lamina propria lymphocytes, conventional dendritic cells, and enterocytes. As compared to chow-fed mice, a high-fat high-sucrose (HFHS) "Western" diet resulted in the accumulation of specific immune cell populations and marked changes to enterocytes nutrient absorption function. Utilizing ligand-receptor analysis, we profiled high-resolution intestine interaction networks across all immune cell and epithelial structural cell types in mice fed chow or HFHS diets. These results revealed novel interactions and communication hubs among intestinal cells, and their potential roles in local as well as systemic inflammation.
Collapse
Affiliation(s)
- Yu-Chen Wang
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Yang Cao
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Zhiqiang Zhou
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles , Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, Los Angeles , Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, the David Geffen School of Medicine, University of California, Los Angeles , Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles , Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, Los Angeles , Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles , Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA , Los Angeles, CA, USA
| |
Collapse
|
12
|
MacPherson KP, Eidson LN, Houser MC, Weiss BE, Gollihue JL, Herrick MK, de Sousa Rodrigues ME, Sniffen L, Weekman EM, Hamilton AM, Kelly SD, Oliver DL, Yang Y, Chang J, Sampson TR, Norris CM, Tansey MG. Soluble TNF mediates amyloid-independent, diet-induced alterations to immune and neuronal functions in an Alzheimer's disease mouse model. Front Cell Neurosci 2023; 17:895017. [PMID: 37006470 PMCID: PMC10052573 DOI: 10.3389/fncel.2023.895017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 01/20/2023] [Indexed: 03/17/2023] Open
Abstract
Introduction: Increasing evidence indicates that neurodegenerative diseases, including Alzheimer's disease (AD), are a product of gene-by-environment interplay. The immune system is a major contributor mediating these interactions. Signaling between peripheral immune cells and those within the microvasculature and meninges of the central nervous system (CNS), at the blood-brain barrier, and in the gut likely plays an important role in AD. The cytokine tumor necrosis factor (TNF) is elevated in AD patients, regulates brain and gut barrier permeability, and is produced by central and peripheral immune cells. Our group previously reported that soluble TNF (sTNF) modulates cytokine and chemokine cascades that regulate peripheral immune cell traffic to the brain in young 5xFAD female mice, and in separate studies that a diet high in fat and sugar (HFHS) dysregulates signaling pathways that trigger sTNF-dependent immune and metabolic responses that can result in metabolic syndrome, which is a risk factor for AD. We hypothesized that sTNF is a key mediator of peripheral immune cell contributions to gene-by-environment interactions to AD-like pathology, metabolic dysfunction, and diet-induced gut dysbiosis. Methods: Female 5xFAD mice were subjected to HFHS diet for 2 months and then given XPro1595 to inhibit sTNF for the last month or saline vehicle. We quantified immune cell profiles by multi-color flow cytometry on cells isolated from brain and blood; metabolic, immune, and inflammatory mRNA and protein marker biochemical and immunhistological analyses, gut microbiome, and electrophysiology in brain slices were also performed. Results: Here, we show that selective inhibition of sTNF signaling via the biologic XPro1595 modulates the effects of an HFHS diet in 5xFAD mice on peripheral and central immune profiles including CNS-associated CD8+ T cells, the composition of gut microbiota, and long-term potentiation deficits. Discussion: Obesogenic diet induces immune and neuronal dysfunction in 5xFAD mice and sTNF inhibition mitigates its effects. A clinical trial in subjects at risk for AD due to genetic predisposition and underlying inflammation associated with peripheral inflammatory co-morbidities will be needed to investigate the extent to which these findings translate to the clinic.
Collapse
Affiliation(s)
- Kathryn P. MacPherson
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Lori N. Eidson
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Madelyn C. Houser
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Blaine E. Weiss
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jenna L. Gollihue
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Mary K. Herrick
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, FL, United States
| | - Maria Elizabeth de Sousa Rodrigues
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Lindsey Sniffen
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Erica M. Weekman
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Adam M. Hamilton
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Sean D. Kelly
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Danielle L. Oliver
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Yuan Yang
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jianjun Chang
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Timothy R. Sampson
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Christopher M. Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Malú Gámez Tansey
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, The University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
13
|
Qiao B, Li X, Peng M, Hui H, Tan Z. Alteration of intestinal mucosal microbiota in mice with Chinese dampness-heat syndrom diarrhea by improper diet combined with high temperature and humidity environments. Front Cell Infect Microbiol 2023; 12:1096202. [PMID: 36683693 PMCID: PMC9845886 DOI: 10.3389/fcimb.2022.1096202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Environment, diet, and emotion may trigger diarrhea, but the mechanism is unclear. Dietary habits or environmental factors affect the composition of gut microbiota. This study aimed to investigate the effects of improper diet combined with high humidity and temperature (HTH) environment on the intestinal mucosal microbiota. MATERIALS AND METHODS Kunming mice were randomly assigned to two equal groups of five mice, namely the control (ccm) group and the model (cmm) group. Diarrhea mice with dampness-heat (DSH) were established by improper diet combined with HTH environments. We used 16S rRNA gene amplicon sequencing to analyze the characteristics of intestinal mucosal microbiota and the interaction relationship of function. RESULTS Our study shows that the intestinal mucosal microbiota of mice changed significantly after an improper diet combined with the HTH environments. The abundance of Fusobacteria and Haemophilus increased dramatically in the cmm group compared to the ccm group (P<0.05). And the abundance of Firmicutes, Lactobacillus, and Lonsdalea was significantly decreased in the cmm group (P<0.05). According to the functional predictive analysis, we found that Lactobacillus showed a significant negative correlation with Protein export, Homologous recombination, Phenylalanine, tyrosine, tryptophan biosynthesis, Citrate cycle, and Lipoic acid metabolism. CONCLUSION Diarrhea with DSH constructed under improper diet and HTH environment may be related to Lactobacillus and Haemophilus. And long-term consumption of improper diet and the HTH environment may affect metabolism.
Collapse
Affiliation(s)
- Bo Qiao
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoya Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Maijiao Peng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Huaying Hui
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Zhoujin Tan
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
14
|
Abstract
The diet and gut microbiota have been extensively interrogated as a fuel for gut inflammation in inflammatory bowel diseases (IBDs) in the last few years. Here, we review how specific nutrients, typically enriched in a Western diet, instigate or deteriorate experimental gut inflammation in a genetically susceptible host and we discuss microbiota-dependent and independent mechanisms. We depict the study landscape of nutritional trials in paediatric and adult IBD and delineate common grounds for dietary advice. Conclusively, the diet reflects a critical rheostat of microbial dysbiosis and gut inflammation in IBD. Dietary restriction by exclusive enteral nutrition, with or without a specific exclusion diet, is effectively treating paediatric Crohn's disease, while adult IBD trials are less conclusive. Insights into molecular mechanisms of nutritional therapy will change the perception of IBD and will allow us to enter the era of precision nutrition. To achieve this, we discuss the need for carefully designed nutritional trials with scientific rigour comparable to medical trials, which also requires action from stake holders. Establishing evidence-based dietary therapy for IBD does not only hold promise to avoid long-term immunosuppression, but to provide a widely accessible therapy at low cost. Identification of dietary culprits disturbing gut health also bears the potential to prevent IBD and allows informed decision making in food politics.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Medicine I, Gastroenterology, Hepatology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
15
|
Srour B, Kordahi MC, Bonazzi E, Deschasaux-Tanguy M, Touvier M, Chassaing B. Ultra-processed foods and human health: from epidemiological evidence to mechanistic insights. Lancet Gastroenterol Hepatol 2022; 7:1128-1140. [PMID: 35952706 DOI: 10.1016/s2468-1253(22)00169-8] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
Epidemiological studies have suggested a role for ultra-processed foods in numerous chronic inflammatory diseases such as inflammatory bowel diseases and metabolic syndrome. Preclinical and clinical studies are accumulating to better decipher the effects of various aspects of food processing and formulation on the aetiology of chronic, debilitating inflammatory diseases. In this Review, we provide an overview of the current data that highlight an association between ultra-processed food consumption and various chronic diseases, with a focus on epidemiological evidence and mechanistic insights involving the intestinal microbiota.
Collapse
Affiliation(s)
- Bernard Srour
- Sorbonne Paris Nord University, INSERM U1153, INRAE U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, Centre of Research in Epidemiology and Statistics, Université Paris Cité, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France
| | - Melissa C Kordahi
- INSERM U1016, Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France
| | - Erica Bonazzi
- INSERM U1016, Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France
| | - Mélanie Deschasaux-Tanguy
- Sorbonne Paris Nord University, INSERM U1153, INRAE U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, Centre of Research in Epidemiology and Statistics, Université Paris Cité, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France
| | - Mathilde Touvier
- Sorbonne Paris Nord University, INSERM U1153, INRAE U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, Centre of Research in Epidemiology and Statistics, Université Paris Cité, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France
| | - Benoit Chassaing
- INSERM U1016, Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France; NACRe Network-Nutrition and Cancer Research Network, Jouy-en-Josas, France.
| |
Collapse
|
16
|
Adolph TE, Meyer M, Schwärzler J, Mayr L, Grabherr F, Tilg H. The metabolic nature of inflammatory bowel diseases. Nat Rev Gastroenterol Hepatol 2022; 19:753-767. [PMID: 35906289 DOI: 10.1038/s41575-022-00658-y] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 02/06/2023]
Abstract
Crohn's disease and ulcerative colitis, phenotypically comprising a spectrum of inflammatory bowel diseases (IBDs), spread globally during the westernization of lifestyle and dietary habits over the past few decades. Here, we review experimental and clinical evidence for the metabolic nature of gut inflammation in IBD and delineate distinct parallels to the inflammatory state in metabolic diseases. Experimental evidence indicates that excessive intake of specific macronutrients in a Western diet fuels an inflammatory response in the gut by exploiting sensors of innate immunity and perturbation of gut microbial metabolism. Genetic IBD risk partly affects metabolism and stress signalling of innate immunity, and immunometabolism controls susceptibility to gut inflammation. Epidemiological and clinical studies indicate that specific nutrients in the Western diet pose a risk for the development of IBD and a poor disease course. Translational studies in IBD indicate perturbation of energy metabolism in immune cells and perturbation of gut microbial metabolism, which can be shaped by diet. In turn, dietary restriction by exclusive enteral nutrition induces remission in patients with IBD. Collectively, these studies support a metabolic underpinning of gut inflammation in IBD as described for metabolic inflammation in obesity and related disorders.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
17
|
Abstract
Changes in the composition of the gut microbiota are associated with many human diseases. So far, however, we have failed to define homeostasis or dysbiosis by the presence or absence of specific microbial species. The composition and function of the adult gut microbiota is governed by diet and host factors that regulate and direct microbial growth. The host delivers oxygen and nitrate to the lumen of the small intestine, which selects for bacteria that use respiration for energy production. In the colon, by contrast, the host limits the availability of oxygen and nitrate, which results in a bacterial community that specializes in fermentation for growth. Although diet influences microbiota composition, a poor diet weakens host control mechanisms that regulate the microbiota. Hence, quantifying host parameters that control microbial growth could help define homeostasis or dysbiosis and could offer alternative strategies to remediate dysbiosis.
Collapse
Affiliation(s)
- Jee-Yon Lee
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA 95616, USA
| |
Collapse
|
18
|
Coleman MJ, Espino LM, Lebensohn H, Zimkute MV, Yaghooti N, Ling CL, Gross JM, Listwan N, Cano S, Garcia V, Lovato DM, Tigert SL, Jones DR, Gullapalli RR, Rakov NE, Torrazza Perez EG, Castillo EF. Individuals with Metabolic Syndrome Show Altered Fecal Lipidomic Profiles with No Signs of Intestinal Inflammation or Increased Intestinal Permeability. Metabolites 2022; 12:431. [PMID: 35629938 PMCID: PMC9143200 DOI: 10.3390/metabo12050431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Metabolic Syndrome (MetS) is a clinical diagnosis where patients exhibit three out of the five risk factors: hypertriglyceridemia, low high-density lipoprotein (HDL) cholesterol, hyperglycemia, elevated blood pressure, or increased abdominal obesity. MetS arises due to dysregulated metabolic pathways that culminate with insulin resistance and put individuals at risk to develop various comorbidities with far-reaching medical consequences such as non-alcoholic fatty liver disease (NAFLD) and cardiovascular disease. As it stands, the exact pathogenesis of MetS as well as the involvement of the gastrointestinal tract in MetS is not fully understood. Our study aimed to evaluate intestinal health in human subjects with MetS. METHODS We examined MetS risk factors in individuals through body measurements and clinical and biochemical blood analysis. To evaluate intestinal health, gut inflammation was measured by fecal calprotectin, intestinal permeability through the lactulose-mannitol test, and utilized fecal metabolomics to examine alterations in the host-microbiota gut metabolism. RESULTS No signs of intestinal inflammation or increased intestinal permeability were observed in the MetS group compared to our control group. However, we found a significant increase in 417 lipid features of the gut lipidome in our MetS cohort. An identified fecal lipid, diacyl-glycerophosphocholine, showed a strong correlation with several MetS risk factors. Although our MetS cohort showed no signs of intestinal inflammation, they presented with increased levels of serum TNFα that also correlated with increasing triglyceride and fecal diacyl-glycerophosphocholine levels and decreasing HDL cholesterol levels. CONCLUSION Taken together, our main results show that MetS subjects showed major alterations in fecal lipid profiles suggesting alterations in the intestinal host-microbiota metabolism that may arise before concrete signs of gut inflammation or intestinal permeability become apparent. Lastly, we posit that fecal metabolomics could serve as a non-invasive, accurate screening method for both MetS and NAFLD.
Collapse
Affiliation(s)
- Mia J. Coleman
- University of New Mexico School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.J.C.); (L.M.E.); (H.L.)
| | - Luis M. Espino
- University of New Mexico School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.J.C.); (L.M.E.); (H.L.)
| | - Hernan Lebensohn
- University of New Mexico School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.J.C.); (L.M.E.); (H.L.)
| | - Marija V. Zimkute
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Negar Yaghooti
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (N.Y.); (C.L.L.); (N.E.R.); (E.G.T.P.)
| | - Christina L. Ling
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (N.Y.); (C.L.L.); (N.E.R.); (E.G.T.P.)
| | - Jessica M. Gross
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Natalia Listwan
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Sandra Cano
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Vanessa Garcia
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Debbie M. Lovato
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Susan L. Tigert
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
| | - Drew R. Jones
- Metabolomics Core Resource Laboratory, New York University Langone Health, New York, NY 10016, USA;
| | - Rama R. Gullapalli
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA;
| | - Neal E. Rakov
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (N.Y.); (C.L.L.); (N.E.R.); (E.G.T.P.)
| | - Euriko G. Torrazza Perez
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (N.Y.); (C.L.L.); (N.E.R.); (E.G.T.P.)
| | - Eliseo F. Castillo
- Clinical and Translational Science Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (M.V.Z.); (J.M.G.); (N.L.); (S.C.); (V.G.); (D.M.L.); (S.L.T.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (N.Y.); (C.L.L.); (N.E.R.); (E.G.T.P.)
| |
Collapse
|