1
|
Zhang H, Tian Y, Xu C, Chen M, Xiang Z, Gu L, Xue H, Xu Q. Crosstalk between gut microbiotas and fatty acid metabolism in colorectal cancer. Cell Death Discov 2025; 11:78. [PMID: 40011436 DOI: 10.1038/s41420-025-02364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy globally and the second leading cause of cancer-related mortality. Its development is a multifactorial and multistage process influenced by a dynamic interplay between gut microbiota, environmental factors, and fatty acid metabolism. Dysbiosis of intestinal microbiota and abnormalities in microbiota-associated metabolites have been implicated in colorectal carcinogenesis, highlighting the pivotal role of microbial and metabolic interactions. Fatty acid metabolism serves as a critical nexus linking dietary patterns with gut microbial activity, significantly impacting intestinal health. In CRC patients, reduced levels of short-chain fatty acids (SCFAs) and SCFA-producing bacteria have been consistently observed. Supplementation with SCFA-producing probiotics has demonstrated tumor-suppressive effects, while therapeutic strategies aimed at modulating SCFA levels have shown potential in enhancing the efficacy of radiation therapy and immunotherapy in both preclinical and clinical settings. This review explores the intricate relationship between gut microbiota, fatty acid metabolism, and CRC, offering insights into the underlying mechanisms and their potential translational applications. Understanding this interplay could pave the way for novel diagnostic, therapeutic, and preventive strategies in the management of CRC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Miaomiao Chen
- Department of Radiology, Huashan Hospital, National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200040, PR China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
2
|
Xia W, Li X, Han R, Liu X. Microbial Champions: The Influence of Gut Microbiota on Athletic Performance via the Gut-Brain Axis. Open Access J Sports Med 2024; 15:209-228. [PMID: 39691802 PMCID: PMC11651067 DOI: 10.2147/oajsm.s485703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024] Open
Abstract
In recent years, exercise has shown a powerful ability to regulate the gut microbiota received with concern. For instance, compared with the sedentary group, high-level athletes showed a different gut microbiota composition and remarkable capability of physiological metabolism. In addition, different diet patterns (eg, high-fat diet, high carbohydrate diet et.al) have different effects on gut microbiota, which can also affect exercise performance. Furthermore, adaptations to exercise also might be influenced by the gut microbiota, due to its important role in the transformation and expenditure of energy obtained from the diet. Therefore, appropriate dietary supplementation is important during exercise. And exploring the mechanisms by which dietary supplements affect exercise performance by modulating gut microbiota is of considerable interest to athletes wishing to achieve health and athletic performance. In this narrative review, the relationship between gut microbiota, dietary supplements, training adaptations and performance is discussed as follows. (i) The effects of the three main nutritional supplements on gut microbiota and athlete fitness. (ii) Strategies for dietary supplements and how they exerted function through gut microbiota alteration based on the gut-brain axis. (iii) Why dietary supplement interventions on gut microbiota should be tailored to different types of exercise. Our work integrates these factors to elucidate how specific nutritional supplements can modulate gut microbiota composition and, consequently, influence training adaptations and performance outcomes, unlike previous literature that often focuses solely on the effects of exercise or diet independently. And provides a comprehensive framework for athletes seeking to optimize their health and performance through a microbiota-centric approach.
Collapse
Affiliation(s)
- Wenrui Xia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaoang Li
- Department of Gastroenterology, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Ruixuan Han
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, People’s Republic of China
| | - Xiaoke Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
3
|
Enache RM, Profir M, Roşu OA, Creţoiu SM, Gaspar BS. The Role of Gut Microbiota in the Onset and Progression of Obesity and Associated Comorbidities. Int J Mol Sci 2024; 25:12321. [PMID: 39596385 PMCID: PMC11595101 DOI: 10.3390/ijms252212321] [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: 10/05/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity, a global public health problem, is constantly increasing, so the concerns in preventing and combating it are increasingly focused on the intestinal microbiota. It was found that the microbiota is different in lean people compared to obese individuals, but the exact mechanisms by which energy homeostasis is influenced are still incompletely known. Numerous studies show the involvement of certain bacterial species in promoting obesity and associated diseases such as diabetes, hypertension, cancer, etc. Our aim is to summarize the main findings regarding the influence of several factors such as lifestyle changes, including diet and bariatric surgery, on the diversity of the gut microbiota in obese individuals. The second purpose of this paper is to investigate the potential effect of various microbiota modulation techniques on ameliorating obesity and its comorbidities. A literature search was conducted using the PubMed database, identifying articles published between 2019 and 2024. Most studies identified suggest that obesity is generally associated with alterations of the gut microbiome such as decreased microbial diversity, an increased Firmicutes-to-Bacteroidetes ratio, and increased SCFAs levels. Our findings also indicate that gut microbiota modulation techniques could represent a novel strategy in treating obesity and related metabolic diseases. Although some mechanisms (e.g., inflammation or hormonal regulation) are already considered a powerful connection between gut microbiota and obesity development, further research is needed to enhance the knowledge on this particular topic.
Collapse
Affiliation(s)
- Robert-Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
4
|
Ioannidis O, Cheva A, Varnalidis I, Koutelidakis I, Papaziogas V, Christidis P, Anestiadou E, Aggelopoulos K, Mantzoros I, Pramateftakis MG, Kotidis E, Driagka B, Aggelopoulos S, Giamarellos-Bourboulis EJ. The Combined Administration of Eicosapentaenoic Acid (EPA) and Gamma-Linolenic Acid (GLA) in Experimentally Induced Colitis: An Experimental Study in Rats. J Clin Med 2024; 13:6661. [PMID: 39597805 PMCID: PMC11594508 DOI: 10.3390/jcm13226661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Ulcerative colitis (UC) is a chronic inflammatory bowel disease with limited effective treatments, prompting the need for investigation of novel therapeutic approaches. Eicosapentaenoic acid (EPA) and gamma-linolenic acid (GLA) have demonstrated potential anti-inflammatory properties, but their combined effects on UC have not been thoroughly investigated. This study aimed to evaluate the effect of the combined administration of EPA and GLA on clinical and histopathologic features of experimental UC models. Methods: Thirty-six male Wistar rats were randomized in three groups (DSS group, Ensure Plus group, and Oxepa group), with twelve rats in each group. Experimental colitis was induced by administrating dextran sulfate sodium (DSS) 8%. The DSS group received tap water, the Ensure Plus group was given a high caloric diet, and the Oxepa group received a special diet containing high levels of EPA and GLA. Disease activity index (DAI) and microscopic activity index (MAI) were measured. Inflammatory markers were calculated both in blood and large intestine, liver, spleen, and lung tissue samples. Neutrophil and macrophage populations were assessed with immunohistochemistry. Results: No significant differences in the DAI index were found between the groups, but the MAI revealed statistically significant differences (p < 0.001). While no significant differences were observed in tumor necrosis factor-alpha (TNF-α) levels, interleukin-17 (IL-17) levels in the large intestine showed statistically significant differences (p = 0.05), with the Ensure Plus and Oxepa groups displaying lower levels compared to the DSS group (p = 0.021 and p = 0.043, respectively). Significant differences in neutrophil infiltration were found in both the large intestine (p < 0.001) and lungs (p = 0.002), with the Oxepa group showing fewer cells. Similarly, significant differences in macrophage infiltration were observed in the large intestine (p = 0.038) and spleen (p < 0.001), with the Oxepa group having lower macrophage counts. Conclusions: In conclusion, the combination of EPA and GLA demonstrates local anti-inflammatory effects and improves the histopathological outcomes in UC.
Collapse
Affiliation(s)
- Orestis Ioannidis
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Angeliki Cheva
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Ioannis Varnalidis
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Ioannis Koutelidakis
- 2nd Department of Surgery, G.Gennimatas General Hospital, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece; (I.K.); (V.P.)
| | - Vasileios Papaziogas
- 2nd Department of Surgery, G.Gennimatas General Hospital, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece; (I.K.); (V.P.)
| | - Panagiotis Christidis
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Elissavet Anestiadou
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Konstantinos Aggelopoulos
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Ioannis Mantzoros
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Manousos George Pramateftakis
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Efstathios Kotidis
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Barbara Driagka
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Stamatios Aggelopoulos
- 4th Department of Surgery, General Hospital “George Papanikolaou”, Aristotle University of Thessaloniki, 57010 Exochi, Greece; (I.V.); (P.C.); (E.A.); (K.A.); (I.M.); (M.G.P.); (E.K.); (B.D.); (S.A.)
| | - Evangelos J. Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens Medical School, “Attikon” Hospital, 12462 Athens, Greece;
| |
Collapse
|
5
|
Zhang K, Zhang Q, Qiu H, Ma Y, Hou N, Zhang J, Kan C, Han F, Sun X, Shi J. The complex link between the gut microbiome and obesity-associated metabolic disorders: Mechanisms and therapeutic opportunities. Heliyon 2024; 10:e37609. [PMID: 39290267 PMCID: PMC11407058 DOI: 10.1016/j.heliyon.2024.e37609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Microbial interactions are widespread and important processes that support the link between disease and microbial ecology. The gut microbiota is a major source of microbial stimuli that can have detrimental or beneficial effects on human health. It is also an endocrine organ that maintains energy homeostasis and host immunity. Obesity is a highly and increasingly prevalent metabolic disease and the leading cause of preventable death worldwide. An imbalance in the gut microbiome is associated with several diseases including obesity-related metabolic disorders. This review summarizes the complex association between the gut microbiome and obesity-associated metabolic diseases and validates the role and mechanisms of ecological dysregulation in the gut in obesity-associated metabolic disorders. Therapies that could potentially alleviate obesity-associated metabolic diseases by modulating the gut microbiota are discussed.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Qi Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yanhui Ma
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
6
|
Esdaille AR, Kuete NK, Anyaeche VI, Kalemoglu E, Kucuk O. The Interplay between Structural Inequality, Allostatic Load, Inflammation, and Cancer in Black Americans: A Narrative Review. Cancers (Basel) 2024; 16:3023. [PMID: 39272881 PMCID: PMC11394332 DOI: 10.3390/cancers16173023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
The impact of racial healthcare disparities has been well documented. Adverse social determinants of health, such as poverty, inadequate housing, and limited access to healthcare, are intricately linked to these disparities and negative health outcomes, highlighting the profound impact that social and economic factors have on individuals' overall well-being. Recent evidence underscores the role of residential location on individual health outcomes. Despite the importance of a healthy lifestyle, regular physical activity, balanced nutrition, and stress management for favorable health outcomes, individuals living in socioeconomically disadvantaged areas may face obstacles in achieving these practices. Adverse living conditions, environmental factors, and systemic biases against Black Americans perpetuate allostatic load. This, compounded by decreased physical activity and limited healthy food options, may contribute to increased oxidative stress and inflammation, fundamental drivers of morbidities such as cardiovascular disease and cancer. Herein, we perform a narrative review of associations between healthcare disparities, chronic stress, allostatic load, inflammation, and cancer in Black Americans, and we discuss potential mechanisms and solutions. Additional research is warranted in the very important area of cancer disparities.
Collapse
Affiliation(s)
- Ashanda R Esdaille
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Division of Urology, Atlanta Veteran's Affairs Medical Center, Decatur, GA 30033, USA
| | - Nelson Kevin Kuete
- Department of Urology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Ecem Kalemoglu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
7
|
Yang C, Xiao J, Xu Z, Wang Z. Gut Microbiota Changes and Its Potential Relations with Thyroid Disorders: From Composition to Therapeutic Targets. Int J Gen Med 2024; 17:3719-3731. [PMID: 39219667 PMCID: PMC11363920 DOI: 10.2147/ijgm.s481183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Composed of over 1200 species of anaerobes and aerobes bacteria along with bacteriophages, viruses, and fungal species, the human gut microbiota (GM) is vital to health, including digestive equilibrium, immunologic, hormonal, and metabolic homeostasis. Micronutrients, usually refer to trace elements (copper, iodine, iron, selenium, zinc) and vitamins (A, C, D, E), interact with the GM to influence host immune metabolism. So far, microbiome studies have revealed an association between disturbances in the microbiota and various pathological disorders, such as anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis, anxiety, depression, early-onset cancers, type 1 diabetes (T1D) and type 2 diabetes (T2D). As common conditions, thyroid diseases, encompassing Graves' disease (GD), Graves' orbitopathy (GO), Hashimoto's thyroiditis (HT), benign nodules, and papillary thyroid cancer (TC), have negative impacts on the health of all populations. Following recent studies, GM might play an integral role in triggering diseases of the thyroid gland. Not only do environmental triggers and genetic predisposing background lead to auto-aggressive damage, involving cellular and humoral networks of the immune system, but the intestinal microbiota interacts with distant organs by signals that may be part of the bacteria themselves or their metabolites. The review aims to describe the current knowledge about the GM in the metabolism of thyroid hormones and the pathogenesis of thyroid diseases and its involvement in the appearance of benign nodules and papillary TC. We further focused on the reciprocal interaction between GM composition and the most used treatment drugs for thyroid disorders. However, the exact etiology has not yet been known. To elucidate more precisely the mechanism for GM involvement in the development of thyroid diseases, future work is needed.
Collapse
Affiliation(s)
- Cai Yang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Jiafeng Xiao
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zibei Xu
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zehong Wang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| |
Collapse
|
8
|
Scarpellini E, Scarlata GGM, Santori V, Scarcella M, Kobyliak N, Abenavoli L. Gut Microbiota, Deranged Immunity, and Hepatocellular Carcinoma. Biomedicines 2024; 12:1797. [PMID: 39200261 PMCID: PMC11351762 DOI: 10.3390/biomedicines12081797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND Liver cancer, particularly hepatocellular carcinoma (HCC), is a significant gastrointestinal disease with a mortality rate as high as nearly 80% within five years. The disease's pathophysiology involves deranged immune responses and bile acid metabolism, with the gut microbiota (GM) playing a crucial role. Recent research highlights the potential of GM in influencing HCC treatment outcomes, especially regarding immune checkpoint inhibitors (ICIs). However, few patients currently benefit from ICIs due to a lack of effective response biomarkers. AIMS AND METHODS This review aimed to explore the literature on HCC treatment issues, focusing on immune response, bile acid metabolism, and GM dysbiosis. This review included studies from PubMed, Medline, and major gastroenterology and hepatology meetings, using keywords like gut microbiota, immune system, liver cancer, and checkpoint inhibitors. RESULTS GM dysbiosis significantly impacts immune response and bile acid metabolism, making it a promising biomarker for ICI response. Modulating GM can enhance ICI treatment efficacy, although more research is needed to confirm its direct therapeutic benefits for HCC. CONCLUSIONS GM dysbiosis is integral to liver cancer pathogenesis and treatment response. Its modulation offers promising therapeutic avenues for improving HCC prognosis and response to immunotherapy.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Translational Research Center in Gastro-Intestinal Disorders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium
| | | | - Valeria Santori
- Gastroenterology Clinic, University of Padua, 35128 Padua, Italy;
| | - Marialaura Scarcella
- Anesthesia, Intensive Care and Nutritional Science—Azienda Ospedaliera “Santa Maria”, Via Tristano di Joannuccio, 05100 Terni, Italy;
| | - Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, 01024 Kyiv, Ukraine;
- Medical Laboratory CSD, 02000 Kyiv, Ukraine
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (G.G.M.S.); (L.A.)
| |
Collapse
|
9
|
Lee S, Portlock T, Le Chatelier E, Garcia-Guevara F, Clasen F, Oñate FP, Pons N, Begum N, Harzandi A, Proffitt C, Rosario D, Vaga S, Park J, von Feilitzen K, Johansson F, Zhang C, Edwards LA, Lombard V, Gauthier F, Steves CJ, Gomez-Cabrero D, Henrissat B, Lee D, Engstrand L, Shawcross DL, Proctor G, Almeida M, Nielsen J, Mardinoglu A, Moyes DL, Ehrlich SD, Uhlen M, Shoaie S. Global compositional and functional states of the human gut microbiome in health and disease. Genome Res 2024; 34:967-978. [PMID: 39038849 PMCID: PMC11293553 DOI: 10.1101/gr.278637.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 06/05/2024] [Indexed: 07/24/2024]
Abstract
The human gut microbiota is of increasing interest, with metagenomics a key tool for analyzing bacterial diversity and functionality in health and disease. Despite increasing efforts to expand microbial gene catalogs and an increasing number of metagenome-assembled genomes, there have been few pan-metagenomic association studies and in-depth functional analyses across different geographies and diseases. Here, we explored 6014 human gut metagenome samples across 19 countries and 23 diseases by performing compositional, functional cluster, and integrative analyses. Using interpreted machine learning classification models and statistical methods, we identified Fusobacterium nucleatum and Anaerostipes hadrus with the highest frequencies, enriched and depleted, respectively, across different disease cohorts. Distinct functional distributions were observed in the gut microbiomes of both westernized and nonwesternized populations. These compositional and functional analyses are presented in the open-access Human Gut Microbiome Atlas, allowing for the exploration of the richness, disease, and regional signatures of the gut microbiota across different cohorts.
Collapse
Affiliation(s)
- Sunjae Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), 61005, Gwangju, Republic of Korea
| | - Theo Portlock
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | | | - Fernando Garcia-Guevara
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Frederick Clasen
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | | | - Nicolas Pons
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Neelu Begum
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Azadeh Harzandi
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Ceri Proffitt
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Dorines Rosario
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Stefania Vaga
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Junseok Park
- Department of Bio and Brain Engineering, KAIST, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Kalle von Feilitzen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Fredric Johansson
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - Lindsey A Edwards
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Vincent Lombard
- INRAE, USC1408 Architecture et Fonction des Macromolécules Biologiques (AFMB), Marseille 13288, France
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille University, Marseille 13288, France
| | - Franck Gauthier
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Claire J Steves
- Department of Twin Research & Genetic Epidemiology, King's College London, London WC2R 2LS, United Kingdom
| | - David Gomez-Cabrero
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Bernard Henrissat
- Department of Biological Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | - Lars Engstrand
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Debbie L Shawcross
- Institute of Liver Studies, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Gordon Proctor
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Mathieu Almeida
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- BioInnovation Institute, DK-2200 Copenhagen N, Denmark
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| | - David L Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom
| | - Stanislav Dusko Ehrlich
- University Paris-Saclay, INRAE, MetaGenoPolis, 78350 Jouy-en-Josas, France
- Department of Clinical and Movement Neurosciences, University College London, London NW3 2PF, United Kingdom
| | - Mathias Uhlen
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden;
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, SE1 9RT, United Kingdom;
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, SE-171 21, Sweden
| |
Collapse
|
10
|
Salo TE, Hakola L, Niinistö S, Takkinen HM, Ahonen S, Puustinen L, Ilonen J, Toppari J, Veijola R, Hyöty H, Knip M, Virtanen SM. Gut Inflammation Markers, Diet, and Risk of Islet Autoimmunity in Finnish Children - A Nested Case-Control Study. J Nutr 2024; 154:2244-2254. [PMID: 38795745 PMCID: PMC11282497 DOI: 10.1016/j.tjnut.2024.05.015] [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: 02/19/2024] [Revised: 04/19/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND Gut dysbiosis and increased intestinal permeability have been reported to precede type 1 diabetes-related autoimmunity. The role of gut inflammation in autoimmunity is not understood. OBJECTIVES This study aimed to assess whether gut inflammation markers are associated with risk of islet autoimmunity and whether diet is associated with gut inflammation markers. METHODS A nested case-control sample of 75 case children with islet autoimmunity and 88 control children was acquired from the Finnish Type 1 Diabetes Prediction and Prevention cohort. Diet was assessed with 3-d food records, and calprotectin and human β-defensin-2 (HBD-2) were analyzed from stool samples at 6 and 12 mo of age. Conditional logistic regression analysis was used in a matched case-control setting to assess risk of autoimmunity. Analysis of variance, independent samples t test, and a general linear model were used in secondary analyses to test associations of background characteristics and dietary factors with inflammation markers. RESULTS In unadjusted analyses, calprotectin was not associated with risk of islet autoimmunity, whereas HBD-2 in the middle (odds ratio [OR]: 3.23; 95% confidence interval [CI]: 1.03, 10.08) or highest tertile (OR: 3.02; 95% CI: 1.05, 8.69) in comparison to the lowest at 12 mo of age showed borderline association (P-trend = 0.063) with higher risk of islet autoimmunity. Excluding children with cow milk allergy in sensitivity analyses strengthened the association of HBD-2 with islet autoimmunity, whereas adjusting for dietary factors and maternal education weakened it. At age 12 mo, higher fat intake was associated with higher HBD-2 (β: 0.219; 95% CI: 0.110, 0.328) and higher intake of dietary fiber (β: -0.294; 95% CI: -0.510, -0.078), magnesium (β: -0.036; 95% CI: -0.059, -0.014), and potassium (β: -0.003; 95% CI: -0.005, -0.001) with lower HBD-2. CONCLUSIONS Higher HBD-2 in infancy may be associated with higher risk of islet autoimmunity. Dietary factors play a role in gut inflammatory status.
Collapse
Affiliation(s)
- Tuuli Ei Salo
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland.
| | - Leena Hakola
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Sari Niinistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Hanna-Mari Takkinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Suvi Ahonen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Leena Puustinen
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, and Centre for Population Health Research, University of Turku, Turku, Finland; Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab laboratories, Tampere, Finland
| | - Mikael Knip
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Suvi M Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
11
|
Pimenta AI, Bernardino RM, Pereira IAC. Role of sulfidogenic members of the gut microbiota in human disease. Adv Microb Physiol 2024; 85:145-200. [PMID: 39059820 DOI: 10.1016/bs.ampbs.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The human gut flora comprises a dynamic network of bacterial species that coexist in a finely tuned equilibrium. The interaction with intestinal bacteria profoundly influences the host's development, metabolism, immunity, and overall health. Furthermore, dysbiosis, a disruption of the gut microbiota, can induce a variety of diseases, not exclusively associated with the intestinal tract. The increased consumption of animal protein, high-fat and high-sugar diets in Western countries has been implicated in the rise of chronic and inflammatory illnesses associated with dysbiosis. In particular, this diet leads to the overgrowth of sulfide-producing bacteria, known as sulfidogenic bacteria, which has been linked to inflammatory bowel diseases and colorectal cancer, among other disorders. Sulfidogenic bacteria include sulfate-reducing bacteria (Desulfovibrio spp.) and Bilophila wadsworthia among others, which convert organic and inorganic sulfur compounds to sulfide through the dissimilatory sulfite reduction pathway. At high concentrations, sulfide is cytotoxic and disrupts the integrity of the intestinal epithelium and mucus barrier, triggering inflammation. Besides producing sulfide, B. wadsworthia has revealed significant pathogenic potential, demonstrated in the ability to cause infection, adhere to intestinal cells, promote inflammation, and compromise the integrity of the colonic mucus layer. This review delves into the mechanisms by which taurine and sulfide-driven gut dysbiosis contribute to the pathogenesis of sulfidogenic bacteria, and discusses the role of these gut microbes, particularly B. wadsworthia, in human diseases.
Collapse
Affiliation(s)
- Andreia I Pimenta
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Raquel M Bernardino
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A C Pereira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
12
|
Mei EH, Yao C, Chen YN, Nan SX, Qi SC. Multifunctional role of oral bacteria in the progression of non-alcoholic fatty liver disease. World J Hepatol 2024; 16:688-702. [PMID: 38818294 PMCID: PMC11135273 DOI: 10.4254/wjh.v16.i5.688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 04/07/2024] [Indexed: 05/22/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver disorders of varying severity, ultimately leading to fibrosis. This spectrum primarily consists of NAFL and non-alcoholic steatohepatitis. The pathogenesis of NAFLD is closely associated with disturbances in the gut microbiota and impairment of the intestinal barrier. Non-gut commensal flora, particularly bacteria, play a pivotal role in the progression of NAFLD. Notably, Porphyromonas gingivalis, a principal bacterium involved in periodontitis, is known to facilitate lipid accumulation, augment immune responses, and induce insulin resistance, thereby exacerbating fibrosis in cases of periodontitis-associated NAFLD. The influence of oral microbiota on NAFLD via the "oral-gut-liver" axis is gaining recognition, offering a novel perspective for NAFLD management through microbial imbalance correction. This review endeavors to encapsulate the intricate roles of oral bacteria in NAFLD and explore underlying mechanisms, emphasizing microbial control strategies as a viable therapeutic avenue for NAFLD.
Collapse
Affiliation(s)
- En-Hua Mei
- Shanghai Medical College, Fudan University, Shanghai 200000, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Chao Yao
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Yi-Nan Chen
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Shun-Xue Nan
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Sheng-Cai Qi
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China.
| |
Collapse
|
13
|
Sant' Ana CT, Verediano TA, Grancieri M, Lopes Toledo RC, Costa NMB, Martino HSD, Barros FARD. Macauba ( Acrocomia aculeata) pulp oil has the potential to enhance the intestinal barrier morphology, goblet cell proliferation and gut microbiota composition in mice fed a high-fat diet. Br J Nutr 2024; 131:987-996. [PMID: 37955051 DOI: 10.1017/s0007114523002623] [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] [Indexed: 11/14/2023]
Abstract
Macauba (Acrocomia aculeata) is a palm tree native from Brazil, whose pulp is rich in oil that has a high content of oleic acid and carotenoids. Macauba pulp oil can bring health benefits due to its bioactive compounds; however, its effects on gut health are unknown. Thus, the objective of this study was to evaluate the effect of macauba pulp oil on the intestinal health in mice fed a high-fat (HF) diet. Male C57BL1/6 mice were randomly divided into three groups (10 animals/group): control diet, HF diet and HF diet with 4 % of macauba pulp oil (HFM). Concentration of short-chain fatty acids (SCFA), faecal pH and histomorphometric analysis of the colon were performed. Content of colon samples was used on microbiome analysis using 16S rRNA amplicon sequencing. Animals from the HFM group had higher butyric acid content and goblet cells number, greater circular and longitudinal muscle layer and higher α-diversity compared with the HF group. Moreover, consumption of MPO reduced Desulfobacterota phylum, Ruminococcaceae, Oscillospiraceae, Prevotellaceae, Bifidobacteriaceae family, Faecalibacterium, Prevotella, Ruminococcus and Enterorhabdus genus. Therefore, macauba pulp oil was able to modulate the gut microbiota and enhance intestinal barrier morphology, showing preventive effects on gut dysbiosis in mice fed a HF diet.
Collapse
Affiliation(s)
- Cíntia Tomaz Sant' Ana
- Department of Food Technology, Federal University of Viçosa, Viçosa, MG36570-000, Brazil
| | | | - Mariana Grancieri
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, MG, Brazil
| | | | | | | | | |
Collapse
|
14
|
Hernández-Martín M, Bocanegra A, Garcimartín A, Issa JÁ, Redondo-Castillejo R, Macho-González A, Benedí J, Sánchez Muniz FJ, López-Oliva ME. Analysis of immunohistomorphological changes in the colonic mucosa in a high-saturated fat and high-cholesterol fed streptozotocin/nicotinamide diabetic rat model. Methods Cell Biol 2024; 185:165-195. [PMID: 38556447 DOI: 10.1016/bs.mcb.2024.02.010] [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] [Indexed: 04/02/2024]
Abstract
The mucosal surface of gastrointestinal tract is lined with epithelial cells that establish an effective barrier between the lumen and internal environment through intercellular junctions, preventing the passage of potentially harmful substances. The "intestinal barrier function" consist of a defensive system that prevent the passage of antigens, toxins, and microbial products, while maintains the correct development of the epithelial barrier, the immune system and the acquisition of tolerance toward dietary antigens and intestinal microbiota. Intestinal morphology changes subsequent to nutritional variations, stress, aging or diseases, which can also affect the composition of the microbiota, altering the homeostasis of the intestine. A growing body of evidence suggests that alterations in intestinal barrier function favor the development of exaggerated immune responses, leading to metabolic endotoxemia, which seems to be the origin of many chronic metabolic diseases such as type 2 diabetes mellitus (T2DM). Although the mechanisms are still unknown, the interaction between dietary patterns, gut microbiota, intestinal mucosa, and metabolic inflammation seems to be a key factor for the development of T2DM, among other diseases. This chapter details the different techniques that allow evaluating the morphological and molecular alterations that lead of the intestinal barrier dysfunction in a T2DM experimental model. To induce both diabetic metabolic disturbances and gut barrier disruption, Wistar rats were fed a high-saturated fat and high-cholesterol diet and received a single dose of streptozotocin/nicotinamide. This animal model may contribute to clarify the understanding of the role of intestinal barrier dysfunction on the late-stage T2DM etiology.
Collapse
Affiliation(s)
- Marina Hernández-Martín
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Aránzazu Bocanegra
- Department of Pharmacology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Alba Garcimartín
- Department of Pharmacology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Jousef Ángel Issa
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Rocío Redondo-Castillejo
- Department of Pharmacology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Adrián Macho-González
- AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain; Department of Nutrition, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - Juana Benedí
- Department of Pharmacology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain
| | - Francisco José Sánchez Muniz
- AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain; Department of Nutrition, Pharmacy School, Complutense University of Madrid, Madrid, Spain
| | - María Elvira López-Oliva
- Departmental Section of Physiology, Pharmacy School, Complutense University of Madrid, Madrid, Spain; AFUSAN Group, Sanitary Research Institute of the San Carlos Clinical Hospital (IdISSC), Madrid, Spain.
| |
Collapse
|
15
|
Fernandes Q, Inchakalody VP, Bedhiafi T, Mestiri S, Taib N, Uddin S, Merhi M, Dermime S. Chronic inflammation and cancer; the two sides of a coin. Life Sci 2024; 338:122390. [PMID: 38160787 DOI: 10.1016/j.lfs.2023.122390] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
The correlation between chronic inflammation and cancer was initially identified in the 19th century. Biomolecules like interleukins, chemokines, tumor necrosis factors, growth factors, and adhesion molecules, which regulate inflammation, are recognized contributors to neoplastic transformation through various mechanisms, including oncogenic mutations, resistance to apoptosis, and adaptive responses like angiogenesis. This review aims to establish connections between the intricate and complex mechanisms of chronic inflammation and cancer. We illuminate implicit signaling mechanisms that drive the association between chronic inflammation and the initiation/progression of cancer, exploring potential impacts on other diseases. Additionally, we discuss the modalities of currently available therapeutic options for chronic inflammation and cancer, emphasizing the dual nature of such therapies. A thorough understanding of the molecular basis of chronic inflammation is crucial for developing novel approaches in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Varghese Philipose Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Takwa Bedhiafi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
16
|
Petito-da-Silva TI, Villardi FM, Penna-de-Carvalho A, Mandarim-de-Lacerda CA, Souza-Mello V, Barbosa-da-Silva S. An Intestinal FXR Agonist Mitigates Dysbiosis, Intestinal Tight Junctions, and Inflammation in High-Fat Diet-Fed Mice. Mol Nutr Food Res 2024; 68:e2300148. [PMID: 38085111 DOI: 10.1002/mnfr.202300148] [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: 03/15/2023] [Revised: 10/23/2023] [Indexed: 03/01/2024]
Abstract
SCOPE To analyze the effects of fexaramine (FEX), as an intestinal FXR agonist, on the modulation of the intestinal microbiota and ileum of mice fed a high-fat (HF) diet. METHODS AND RESULTS Three-month-old C57Bl/6 male mice are divided into two groups and received a control (C, 10% of energy from lipids) or HF (50% of energy from lipids) diet for 12 weeks. They are subdivided into the C, C + FEX, HF, and HF + FEX groups. FEX is administered (FEX-5 mg kg-1 ) via orogastric gavage for three weeks. Body mass (BM), glucose metabolism, qPCR 16S rRNA gene expression, and ileum gene expression, bile acids (BAs), tight junctions (TJs), and incretin are analyzed. FEX reduces BM and glucose intolerance, reduces plasma lipid concentrations and the Firmicutes/Bacteroidetes ratio, increases the Lactobacillus sp. and Prevotella sp. abundance, and reduces the Escherichia coli abundance. Consequently, the ileal gene expression of Fxr-Fgf15, Tgr5-Glp1, and Cldn-Ocldn-Zo1 is increased, and Tlr4-Il6-Il1beta is decreased. CONCLUSION FEX stimulates intestinal FXR and improves dysbiosis, intestinal TJs, and the release of incretins, mitigating glucose intolerance and BM increases induced by an HF diet. However, FEX results in glucose intolerance, insulin resistance, and reduces intestinal TJs in a control group, thus demonstrating limitations to this dietary model.
Collapse
Affiliation(s)
- Tamiris Ingrid Petito-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe Missiba Villardi
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Penna-de-Carvalho
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Engin AB, Engin A. The Checkpoints of Intestinal Fat Absorption in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:73-95. [PMID: 39287849 DOI: 10.1007/978-3-031-63657-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In this chapter, intestinal lipid transport, which plays a central role in fat homeostasis and the development of obesity in addition to the mechanisms of fatty acids and monoacylglycerol absorption in the intestinal lumen and reassembly of these within the enterocyte was described. A part of the resynthesized triglycerides (triacylglycerols; TAG) is repackaged in the intestine to form the hydrophobic core of chylomicrons (CMs). These are delivered as metabolic fuels, essential fatty acids, and other lipid-soluble nutrients, from enterocytes to the peripheral tissues following detachment from the endoplasmic reticulum membrane. Moreover, the attitudes of multiple receptor functions in dietary lipid uptake, synthesis, and transport are highlighted. Additionally, intestinal fatty acid binding proteins (FABPs), which increase the cytosolic flux of fatty acids via intermembrane transfer in enterocytes, and the functions of checkpoints for receptor-mediated fatty acid signaling are debated. The importance of the balance between storage and secretion of dietary fat by enterocytes in determining the physiological fate of dietary fat, including regulation of blood lipid concentrations and energy balance, is mentioned. Consequently, promising checkpoints regarding how intestinal fat processing affects lipid homeostatic mechanisms and lipid stores in the body and the prevention of obesity-lipotoxicity due to excessive intestinal lipid absorption are evaluated. In this context, dietary TAG digestion, pharmacological inhibition of TAG hydrolysis, the regulation of long-chain fatty acid uptake traffic into adipocytes, intracellular TAG resynthesis, the enlargement of cytoplasmic lipid droplets in enterocytes and constitutional alteration of their proteome, CD36-mediated conversion of diet-derived fatty acid into cellular lipid messengers and their functions are discussed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
18
|
Rajapakse J, Khatiwada S, Akon AC, Yu KL, Shen S, Zekry A. Unveiling the complex relationship between gut microbiota and liver cancer: opportunities for novel therapeutic interventions. Gut Microbes 2023; 15:2240031. [PMID: 37615334 PMCID: PMC10454000 DOI: 10.1080/19490976.2023.2240031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has been linked to the gut microbiota, with recent studies revealing the potential of gut-generated responses to influence several arms of the immune responses relevant to HCC formation. The pro- or anti-tumor effects of specific bacterial strains or gut microbiota-related metabolites, such as bile acids and short-chain fatty acids, have been highlighted in many human and animal studies. The critical role of the gut microbiota in HCC development has spurred interest in modulating the gut microbiota through dietary interventions, probiotics, and fecal microbiota transplantation as a potential strategy to improve liver cancer outcomes. Encouragingly, preclinical and clinical studies have demonstrated that modulation of the gut microbiota can ameliorate liver function, reduce inflammation, and inhibit liver tumor growth, underscoring the potential of this approach to improve HCC outcomes. As research continues to unravel the complex and dynamic mechanisms underlying the gut-liver axis, the development of safe and effective interventions to target this pathway for liver cancer prevention and treatment appears to be on the horizon, heralding a significant advance in our ongoing efforts to combat this devastating disease.
Collapse
Affiliation(s)
- Jayashi Rajapakse
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales (UNSW), Sydney, Australia
| | - Saroj Khatiwada
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales (UNSW), Sydney, Australia
| | - Anna Camille Akon
- St George Hospital, Gastroenterology and Hepatology Department, Sydney, Australia
| | - Kin Lam Yu
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales (UNSW), Sydney, Australia
| | - Sj Shen
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales (UNSW), Sydney, Australia
| | - Amany Zekry
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales (UNSW), Sydney, Australia
- St George Hospital, Gastroenterology and Hepatology Department, Sydney, Australia
| |
Collapse
|
19
|
Shateri Z, Makhtoomi M, Mansouri F, Rajabzadeh-Dehkordi M, Nouri M, Rashidkhani B. The association between empirical dietary inflammatory pattern and colorectal cancer risk: a case-control study. BMC Nutr 2023; 9:136. [PMID: 37996874 PMCID: PMC10666418 DOI: 10.1186/s40795-023-00797-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer in Iran. Inflammation plays an essential role in developing CRC. A dietary pattern called the empirical dietary inflammatory pattern (EDIP) has recently been designed based on the inflammatory potential of the diet. Therefore, the present study aimed to investigate the impact of EDIP on the risk of CRC. METHODS The current case-control study was conducted on 142 controls and 71 CRC cases in three general hospitals and Hospital Cancer Organization in Tehran, Iran. We calculated EDIP by a semi-quantitative food frequency questionnaire. The association between EDIP and CRC were evaluated by logistic regression. The level of significance was p < 0.05. RESULTS The results revealed that people who were in the highest tertile of the EDIP had higher odds of CRC (in the adjusted model: odds ratio (OR) = 3.74; 95% confidence interval (CI): 1.38-10.14; P = 0.011). CONCLUSION The present study demonstrated the potential role of dietary-induced inflammation in developing CRC. In the current study, an increase in the intake of red meat, processed meats, and refined grains was observed in the higher EDIP tertiles compared to the lower tertiles. Consequently, to decrease the risk of CRC, it is recommended to reduce the consumption of these foods.
Collapse
Affiliation(s)
- Zainab Shateri
- Department of Nutrition and Biochemistry, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Maede Makhtoomi
- Students' Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Mansouri
- Students' Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Rajabzadeh-Dehkordi
- Students' Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehran Nouri
- Students' Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahram Rashidkhani
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Li J, Ma X, Luo L, Tang D, Zhang L. The What and Who of Dietary Lignans in Human Health: Special Attention to Estrogen Effects and Safety Evaluation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16419-16434. [PMID: 37870451 DOI: 10.1021/acs.jafc.3c02680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Lignans are a group of phenolic compounds found in plant-based diets. The human body can obtain lignans through diet, which are then metabolized into enterolignans. The enterolignans have been linked to several health benefits, including anticancer, anti-inflammatory, antioxidant effects, and estrogen effects. This review explores the relationship between the estrogenic effects of lignans and health. This review not only considers the estrogen-like activity of lignans but also discusses the safe dosage of lignans at different life stages. In addition, this review also identified other types of bioactive compounds that can act synergistically with lignans to promote health. Studies have shown that lignan administration during pregnancy and lactation reduces the risk of breast cancer in offspring. Further studies are needed to investigate the estrogenic safety effects of lignan on pregnant women and children. Whether lignans combine with other nutrients in complex food substrates to produce synergistic effects remains to be investigated. This review provides a basis for future studies on the safe dose of lignan and recommended dietary intake of lignan. We believe that the acquired as discussed here has implications for developing dietary therapies that can promote host nutrition and modulate estrogenic diseases.
Collapse
Affiliation(s)
- Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xiaoyang Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Lianzhong Luo
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources, Xiamen Medical College, Xiamen 361023, China
| | - Danqing Tang
- The School of Foreign Languages of Jimei University, Xiamen 361021, China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
21
|
Li Z, Cui R, Wang YB, Luo YB, Xue PX, Tang QG, Fang MY. Specific gastrointestinal microbiota profiles in Chinese Tan sheep are associated with lauric acid content in muscle. BMC Microbiol 2023; 23:331. [PMID: 37936065 PMCID: PMC10631117 DOI: 10.1186/s12866-023-03079-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/21/2023] [Indexed: 11/09/2023] Open
Abstract
The biological mechanisms underlying meat quality remain unclear. Currently, many studies report that the gastrointestinal microbiota is essential for animal growth and performance. However, it is uncertain which bacterial species are specifically associated with the meat quality traits. In this study, 16S rDNA and metagenomic sequencing were performed to explore the composition and function of microbes in various gastrointestinal segments of Tan sheep and Dorper sheep, as well as the relationship between microbiota and meat quality (specifically, the fatty acid content of the muscle). In the ruminal, duodenal, and colonic microbiome, several bacteria were uniquely identified in respective breeds, including Agrobacterium tumefaciens, Bacteroidales bacterium CF, and several members of the family Oscillospiraceae. The annotation of GO, KEGG, and CAZYme revealed that these different bacterial species were linked to the metabolism of glucose, lipids, and amino acids. Additionally, our findings suggested that 16 microbial species may be essential to the content of fatty acids in the muscle, especially C12:0 (lauric acid). 4 bacterial species, including Achromobacter xylosoxidans, Mageeibacillus indolicus, and Mycobacterium dioxanotrophicus, were positively correlated with C12:0, while 13 bacteria, including Methanobrevibacter millerae, Bacteroidales bacterium CF, and Bacteroides coprosuis were negatively correlated with C12:0. In a word, this study provides a basic data for better understanding the interaction between ruminant gastrointestinal microorganisms and the meat quality traits of hosts.
Collapse
Affiliation(s)
- Zhen Li
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ran Cui
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yu-Bei Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ya-Biao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Peng-Xiang Xue
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qi-Guo Tang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mei-Ying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
22
|
Kazura W, Michalczyk K, Stygar D. The Relationship between the Source of Dietary Animal Fats and Proteins and the Gut Microbiota Condition and Obesity in Humans. Nutrients 2023; 15:3082. [PMID: 37513500 PMCID: PMC10385089 DOI: 10.3390/nu15143082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The relationship between gut microbiota and obesity is well documented in humans and animal models. Dietary factors can change the intestinal microbiota composition and influence obesity development. However, knowledge of how diet, metabolism, and intestinal microbiota interact and modulate energy metabolism and obesity development is still limited. Epidemiological studies show a link between consuming dietary proteins and fats from specific sources and obesity. Animal studies confirm that proteins and fats of different origins differ in their ability to prevent or induce obesity. Protein sources, such as meat, dairy products, vegetables, pulses, and seafood, vary in their amino acid composition. In addition, the type and level of other factors, such as fatty acids or persistent organic pollutants, vary depending on the source of dietary protein. All these factors can modulate the intestinal microbiota composition and, thus, may influence obesity development. This review summarizes selected evidence of how proteins and fats of different origins affect energy efficiency, obesity development, and intestinal microbiota, linking protein and fat-dependent changes in the intestinal microbiota with obesity.
Collapse
Affiliation(s)
- Wojciech Kazura
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
| | - Katarzyna Michalczyk
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
| | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences, Medical University of Silesia, Jordana Street 19, 41-808 Zabrze, Poland
- SLU University Animal Hospital, Swedish University of Agricultural Sciences, SE-750 07 Uppsala, Sweden
| |
Collapse
|
23
|
Pirozzi C, Opallo N, Coretti L, Lama A, Annunziata C, Comella F, Melini S, Buommino E, Mollica MP, Aviello G, Mattace Raso G, Lembo F, Meli R. Alkalihalobacillus clausii (formerly Bacillus clausii) spores lessen antibiotic-induced intestinal injury and reshape gut microbiota composition in mice. Biomed Pharmacother 2023; 163:114860. [PMID: 37196540 DOI: 10.1016/j.biopha.2023.114860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/19/2023] Open
Abstract
The antibiotic-induced intestinal injury (AIJ) is associated with diarrhoea and gastrointestinal discomfort. However, the pathological intestinal mechanisms and related side effects associated with antibiotic use/misuse may be counteracted by probiotics. This study aims to evaluate the effect and the protective mechanisms of a probiotic formulation containing Alkalihalobacillus clausii (formerly Bacillus clausii; BC) spores in an experimental model of AIJ. C57/Bl6J mice were orally challenged with a high dose of ceftriaxone for five days along with BC treatment which lasted up to the 15th day. Our results showed the beneficial effect of the probiotic in preserving colonic integrity and limiting tissue inflammation and immune cell infiltration in AIJ mice. BC increased tight junction expression and regulated the unbalanced production of colonic pro- and anti-inflammatory cytokines, converging toward the full resolution of the intestinal damage. These findings were supported by the histological evaluation of the intestinal mucosa, suggesting a potential restoration of mucus production. Notably, BC treatment increased gene transcription of the secretory products responsible for epithelium repair and mucus synthesis and normalized the expression of antimicrobial peptides involved in immune activation. Reconstruction of complex and diverse gut microbiota in antibiotic-induced dysbiosis was recorded upon BC supplementation. Specifically, the expansion of A. clausii, Prevotella rara and Eubacterium ruminatium drove intestinal microbiota rebalance by primarily impacting Bacteroidota members. Taken together, our data indicate that BC administration alleviates AIJ by multiple converging mechanisms leading to restoring gut integrity and homeostasis and reshaping microbiota composition.
Collapse
Affiliation(s)
- C Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - N Opallo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - L Coretti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - A Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - C Annunziata
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - F Comella
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - S Melini
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - E Buommino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - M P Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - G Aviello
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - G Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy.
| | - F Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - R Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
24
|
Cabello-Olmo M, Oneca M, Urtasun R, Pajares MJ, Goñi S, Riezu-Boj JI, Milagro FI, Ayo J, Encio IJ, Barajas M, Araña M. Pediococcus acidilactici pA1c ® Improves the Beneficial Effects of Metformin Treatment in Type 2 Diabetes by Controlling Glycaemia and Modulating Intestinal Microbiota. Pharmaceutics 2023; 15:pharmaceutics15041203. [PMID: 37111688 PMCID: PMC10143274 DOI: 10.3390/pharmaceutics15041203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic disease, which involves maintained hyperglycemia, mainly due to the development of an insulin resistance process. Metformin administration is the most prescribed treatment for diabetic patients. In a previously published study, we demonstrated that Pediococcus acidilactici pA1c® (pA1c) protects from insulin resistance and body weight gain in HFD-induced diabetic mice. The present work aimed to evaluate the possible beneficial impact of a 16-week administration of pA1c, metformin, or the combination of pA1c and metformin in a T2D HFD-induced mice model. We found that the simultaneous administration of both products attenuated hyperglycemia, increased high-intensity insulin-positive areas in the pancreas and HOMA-β, decreased HOMA-IR and also provided more beneficial effects than metformin treatment (regarding HOMA-IR, serum C-peptide level, liver steatosis or hepatic Fasn expression), and pA1c treatment (regarding body weight or hepatic G6pase expression). The three treatments had a significant impact on fecal microbiota and led to differential composition of commensal bacterial populations. In conclusion, our findings suggest that P. acidilactici pA1c® administration improved metformin beneficial effects as a T2D treatment, and it would be a valuable therapeutic strategy to treat T2D.
Collapse
Affiliation(s)
- Miriam Cabello-Olmo
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - María Oneca
- Genbioma Aplicaciones S.L. Polígono Industrial Noain-Esquíroz, Calle S, Nave 4, 31191 Esquíroz, Spain
| | - Raquel Urtasun
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - María J Pajares
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
- IDISNA Navarra's Health Research Institute, 31008 Pamplona, Spain
| | - Saioa Goñi
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - José I Riezu-Boj
- IDISNA Navarra's Health Research Institute, 31008 Pamplona, Spain
- Center for Nutrition Research, Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Spain
| | - Fermín I Milagro
- IDISNA Navarra's Health Research Institute, 31008 Pamplona, Spain
- Center for Nutrition Research, Department of Nutrition, Food Science and Physiology, University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josune Ayo
- Genbioma Aplicaciones S.L. Polígono Industrial Noain-Esquíroz, Calle S, Nave 4, 31191 Esquíroz, Spain
| | - Ignacio J Encio
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - Miguel Barajas
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| | - Miriam Araña
- Biochemistry Area, Department of Health Science, Public University of Navarre, 31008 Pamplona, Spain
| |
Collapse
|
25
|
Xiong W, Zhang B, Gu Z, Muir J, Dhital S. The microbiota and metabolites during the fermentation of intact plant cells depend on the content of starch, proteins and lipids in the cells. Int J Biol Macromol 2023; 226:965-973. [PMID: 36526066 DOI: 10.1016/j.ijbiomac.2022.12.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/28/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Intact cells, as the smallest unit of whole foods, were isolated from three legume crops and fermented with human faecal inoculum to elucidate the effect of food macro-nutrients compositional difference (starch, proteins and lipids) on in vitro colonic fermentation profiles. After 48 h of fermentation, the highest production of short-chain fatty acids (SCFAs) were observed for the pea cells, abundance in starch (64.9 %, db). In contrast, branch chain fatty acids (BCFAs) were the major metabolites for protein-enriched soybean cells (protein content 56.9 %, db). The peanut cells rich in lipids (49.2 %, db) has the lowest fermentation rate among the three varieties. Correspondingly, pea cells favoured the growth of Bifidobacterium, whereas soybean and peanut cells promoted an abundance of Bacteroides and Shigella, respectively. Furthermore, except the intact pea cells promoting the abundance of butyrate producer Roseburia, a similar fermentation pattern was found between intact and broken cells suggesting that macro-nutrient types, rather than structure, dominate the production of metabolites in colonic fermentation. The findings elucidate how the food compositional difference can modulate the gut microbiome and thus provide the knowledge to design whole food legumes-based functional foods.
Collapse
Affiliation(s)
- Weiyan Xiong
- Department of Chemical and Biological Engineering, Monash University, Clayton Campus, VIC 3800, Australia
| | - Bin Zhang
- Sino-Singapore International Research Institute, Guangzhou 510555, China; School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Zhipeng Gu
- Sino-Singapore International Research Institute, Guangzhou 510555, China; School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jane Muir
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sushil Dhital
- Department of Chemical and Biological Engineering, Monash University, Clayton Campus, VIC 3800, Australia.
| |
Collapse
|
26
|
Effects of a High-Fat Diet on Intestinal and Gonadal Metabolism in Female and Male Sea Cucumber Apostichopus japonicus. BIOLOGY 2023; 12:biology12020212. [PMID: 36829488 PMCID: PMC9953091 DOI: 10.3390/biology12020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
Parental nutrient reserves are directly related to reproductive performance in sea cucumbers. This study focused on the lipid requirements of male and female sea cucumbers Apostichopus japonicus during the reproductive stage and analyzed their physiological responses to a high-fat diet (HFD). The intestinal lipid metabolites and microbiome profile changed significantly in animals fed with the HFD, as given by an upregulation of metabolites related to lipid metabolism and an increase in the predominance of Proteobacteria in the microbiome, respectively. The metabolic responses of male and female sea cucumbers to the HFD differed, which in turn could have triggered sex-related differences in the intestinal microbiome. These results suggest that the lipid content in diets can be differentially adjusted for male and female sea cucumbers to improve nutrition and promote reproduction. This data contributes to a better understanding of the reproductive biology and sex differences of sea cucumbers.
Collapse
|
27
|
Jiang X, Yang Q, Qu H, Chen Y, Zhu S. Endogenous n-3 PUFAs Improve Non-Alcoholic Fatty Liver Disease through FFAR4-Mediated Gut-Liver Crosstalk. Nutrients 2023; 15:nu15030586. [PMID: 36771292 PMCID: PMC9919706 DOI: 10.3390/nu15030586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
The gut-liver axis plays a key role in the development and progression of non-alcoholic fatty liver disease (NAFLD). Due to the complexity and incomplete understanding of the cross-talk between the gut and liver, effective therapeutic targets are largely unknown. Free fatty acid receptors (FFARs) may bridge the cross-talk between the gut and liver. FFAR4 has received considerable attention due to its important role in lipid metabolism. However, the role of FFAR4 in this cross talk in NAFLD remains unclear. In this study, mice with high endogenous n-3 PUFAs but FFAR4 deficiency were generated by crossbreeding Fat-1 and FFAR4 knockout mice. FFAR4 deficiency blocked the protective effects of high endogenous n-3 PUFAs on intestinal barrier dysfunction and hepatic steatosis. In addition, FFAR4 deficiency decreased gut microbiota diversity and enriched Rikenella, Anaerotruncus, and Enterococcus, and reduced Dubosiella, Ruminococcaceae UCG-010, Ruminococcaceae UCG-014, Coriobacteriaceae UCG-002, Faecalibaculum, Ruminococcaceae UCG-009, and Akkermansia. Notably, FFAR4 deficiency co-regulated pantothenic acid and CoA biosynthesis, β-alanine metabolism, and sphingolipid metabolism pathways in the gut and liver, potentially associated with the aggravation of NAFLD. Together, the beneficial effects of n-3 PUFAs on the gut and liver were mediated by FFAR4, providing insights on the role of FFAR4 in the treatment of NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
28
|
Bhat MA, Mishra AK, Tantray JA, Alatawi HA, Saeed M, Rahman S, Jan AT. Gut Microbiota and Cardiovascular System: An Intricate Balance of Health and the Diseased State. Life (Basel) 2022; 12:1986. [PMID: 36556351 PMCID: PMC9780831 DOI: 10.3390/life12121986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Gut microbiota encompasses the resident microflora of the gut. Having an intricate relationship with the host, it plays an important role in regulating physiology and in the maintenance of balance between health and disease. Though dietary habits and the environment play a critical role in shaping the gut, an imbalance (referred to as dysbiosis) serves as a driving factor in the occurrence of different diseases, including cardiovascular disease (CVD). With risk factors of hypertension, diabetes, dyslipidemia, etc., CVD accounts for a large number of deaths among men (32%) and women (35%) worldwide. As gut microbiota is reported to have a direct influence on the risk factors associated with CVDs, this opens up new avenues in exploring the possible role of gut microbiota in regulating the gross physiological aspects along the gut-heart axis. The present study elaborates on different aspects of the gut microbiota and possible interaction with the host towards maintaining a balance between health and the occurrence of CVDs. As the gut microbiota makes regulatory checks for these risk factors, it has a possible role in shaping the gut and, as such, in decreasing the chances of the occurrence of CVDs. With special emphasis on the risk factors for CVDs, this paper includes information on the prominent bacterial species (Firmicutes, Bacteriodetes and others) towards an advance in our understanding of the etiology of CVDs and an exploration of the best possible therapeutic modules for implementation in the treatment of different CVDs along the gut-heart axis.
Collapse
Affiliation(s)
- Mujtaba Aamir Bhat
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Javeed Ahmad Tantray
- Department of Zoology, Central University of Kashmir, Ganderbal 191131, Jammu and Kashmir, India
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail 55476, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur 842001, Bihar, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| |
Collapse
|
29
|
Inflammatory Bowel Disease and Customized Nutritional Intervention Focusing on Gut Microbiome Balance. Nutrients 2022; 14:nu14194117. [PMID: 36235770 PMCID: PMC9572914 DOI: 10.3390/nu14194117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/24/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Inflammatory bowel disease (IBD) represents a chronic relapsing–remitting condition affecting the gastrointestinal system. The specific triggering IBD elements remain unknown: genetic variability, environmental factors, and alterations in the host immune system seem to be involved. An unbalanced diet and subsequent gut dysbiosis are risk factors, too. This review focuses on the description of the impact of pro- and anti-inflammatory food components on IBD, the role of different selected regimes (such as Crohn’s Disease Exclusion Diet, Immunoglobulin Exclusion Diet, Specific Carbohydrate Diet, LOFFLEX Diet, Low FODMAPs Diet, Mediterranean Diet) in the IBD management, and their effects on the gut microbiota (GM) composition and balance. The purpose is to investigate the potential positive action on IBD inflammation, which is associated with the exclusion or addition of certain foods or nutrients, to more consciously customize the nutritional intervention, taking also into account GM fluctuations during both disease flare-up and remission.
Collapse
|
30
|
Bilal M, Ashraf S, Zhao X. Dietary Component-Induced Inflammation and Its Amelioration by Prebiotics, Probiotics, and Synbiotics. Front Nutr 2022; 9:931458. [PMID: 35938108 PMCID: PMC9354043 DOI: 10.3389/fnut.2022.931458] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
A balanced diet with many dietary components maintains immune homeostasis directly by interacting with innate and adaptive immune components or indirectly through gut microbiota and their metabolites. Dietary components may inhibit pro-inflammatory mediators and promote anti-inflammatory functions or vice versa. Western diets with imbalanced dietary components skew the immune balance toward pro-inflammation and induce intestinal inflammation, consequently leading to many intestinal and systemic inflammatory diseases like ulcerative colitis, Crohn's disease, irritable bowel syndrome, cardiovascular problems, obesity, and diabetes. The dietary component-induced inflammation is usually chronic in nature and frequently caused or accompanied by alterations in gut microbiota. Therefore, microbiome-targeted therapies such as probiotics, prebiotics and synbiotics hold great potentials to amend immune dysregulation and gut dysbiosis, preventing and treating intestinal and systemic inflammatory diseases. Probiotics, prebiotics and synbioitcs are progressively being added to foods and beverages, with claims of health benefits. However, the underlining mechanisms of these interventions for preventing and treating dietary component-induced inflammation are still not very clear. In addition, possibly ineffective or negative consequences of some probiotics, prebiotics and synbiotics call for stringent testing and regulation. Here, we will first briefly review inflammation, in terms of its types and the relationship between different dietary components and immune responses. Then, we focus on current knowledge about the direct and indirect effects of probiotics, prebiotics and synbiotics on intestinal and systemic inflammation. Understanding how probiotics, prebiotics and synbiotics modulate the immune system and gut microbiota will improve our strategies for preventing and treating dietary component-induced intestinal inflammation and inflammatory diseases.
Collapse
|
31
|
Wang L, Nong Q, Zhou Y, Sun Y, Chen W, Xie J, Zhu X, Shan T. Changes in Serum Fatty Acid Composition and Metabolome-Microbiome Responses of Heigai Pigs Induced by Dietary N-6/n-3 Polyunsaturated Fatty Acid Ratio. Front Microbiol 2022; 13:917558. [PMID: 35814644 PMCID: PMC9257074 DOI: 10.3389/fmicb.2022.917558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022] Open
Abstract
Changing fatty acid composition is a potential nutritional strategy to shape microbial communities in pigs. However, the effect of different n-6/n-3 polyunsaturated fatty acid (PUFA) ratios on serum fatty acid composition, microbiota, and their metabolites in the intestine of pigs remains unclear. Our study investigated the changes in serum fatty acid composition and metabolome–microbiome responses induced by dietary n-6/n-3 PUFA ratio based on a Heigai-pig model. A total of 54 Heigai finishing pigs (body weight: 71.59 ± 2.16 kg) fed with 3 types of diets (n-6/n-3 PUFA ratios are 8:1, 5:1, and 3:1) were randomly divided into 3 treatments with 6 replications (3 pigs per replication) for 75 days. Results showed that dietary n-6/n-3 PUFA ratio significantly affected biochemical immune indexes including glucose (Glu), triglycerides (TG), total cholesterol (TChol), non-esterified fatty acid (NEFA), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and total thyroxine (TT4), and medium- and long-chain fatty acid composition, especially n-3 PUFA and n-6/n-3 PUFA ratio in the serum. However, no significant effects were found in the SCFAs composition and overall composition of the gut microbiota community. In the low dietary n-6/n-3 PUFA ratio group, the relative abundance of Cellulosilyticum, Bacteroides, and Alloprevotella decreased, Slackia and Sporobacter increased. Based on the metabolomic analysis, dietary n-6/n-3 PUFA ratio altered the metabolome profiles in the colon. Moreover, Pearson’s correlation analysis indicated that differential microbial genera and metabolites induced by different n-6/n-3 PUFA ratio had tight correlations and were correlated with the n-6 PUFA and n-3 PUFA content in longissimus dorsi muscle (LDM) and subcutaneous adipose tissue (SAT). Taken together, these results showed that lower dietary n-6/n-3 PUFA ratio improved serum fatty acid composition and metabolome–microbiome responses of Heigai pigs and may provide a new insight into regulating the metabolism of pigs and further better understanding the crosstalk with host and microbes in pigs.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Qiuyun Nong
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Yanbing Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Ye Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Jintang Xie
- Shandong Chunteng Food Co. Ltd., Zaozhuang, China
| | - Xiaodong Zhu
- Shandong Chunteng Food Co. Ltd., Zaozhuang, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
- *Correspondence: Tizhong Shan,
| |
Collapse
|
32
|
Ye Z, Xu YJ, Liu Y. Different typical dietary lipid consumption affects the bile acid metabolism and the gut microbiota structure: an animal trial using Sprague-Dawley rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3179-3192. [PMID: 34787315 DOI: 10.1002/jsfa.11661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 10/07/2021] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The palm oil (PO), leaf lard oil (LO), rapeseed oil (RO), sunflower oil (SO) and linseed oil (LN) are five of the most typical dietary lipids in most Asian countries. However, their influences on gut health, and the connections between the fatty acid composition, the gut microbiota, and the bile acid metabolism are not fully understood. RESULTS In the present study, results showed that compared with polyunsaturated fatty acid (PUFA)-rich SO and LN, the saturated fatty acid (SFA)-rich and monounsaturated fatty acid (MUFA)-rich PO, LO and RO were more likely to decrease the re-absorption of bile acid in the colon, which was probably caused by their different role in modulating the gut microbiota structure. LO consumption significantly up-regulated the Cyp27a1, FXR and TGR5 gene expression level (P < 0.05). The correlation results suggested that the C18:0 was significantly positive correlated with these three genes, indicating that intake of SFA-rich dietary lipids, especially for the C18:0, could specifically increase the bile acid production by stimulating the bile acid alternative synthesis pathway. Although the bile acid receptor expression in the colon was increased, the re-absorption of bile acid did not show a significant increase (P > 0.05) as compared with other dietary lipids. Moreover, the C18:2-rich SO maintained the bile acid metabolic balance probably by decreasing the Romboutsia, while increasing the Bifidobacterium abundance in the colon. CONCLUSIONS The different dietary lipids showed different effects on the bile acid metabolism, which was probably connected with the alterations in the gut microbiota structure. The present study could provide basic understandings about the influences of the different dietary lipids consumption on gut homeostasis and bile acid metabolism. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhan Ye
- School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, P. R. China
| | - Yong-Jiang Xu
- School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, P. R. China
| | - Yuanfa Liu
- School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
- State Key Laboratory of Food Science and Technology, National Engineering Laboratory for Cereal Fermentation Technology, National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, P. R. China
| |
Collapse
|
33
|
Evaluation of the Effect of Different Dietary Lipid Sources on Dogs’ Faecal Microbial Population and Activities. Animals (Basel) 2022; 12:ani12111368. [PMID: 35681832 PMCID: PMC9179278 DOI: 10.3390/ani12111368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Saturated fatty acids might be a valuable source of energy to guarantee all physiological functions in companion animals. Polyunsaturated fatty acids are essential in several metabolic processes and structural body functions. In this regard, hemp oil can be used as a rich source of polyunsaturated fatty acids in animal diets. In this study, hemp seed oil and swine tallow were added to a commercial canned diet. These high-lipid-content diets (hemp diet: 55.1 g/100 kcal ME; tallow diet: 65.1 g/1000 kcal ME) were compared with one rich in starch. Following the recruitment of 12 dogs, three experimental groups were set up. At 30 days of diet administration, faeces samples were collected from each group to perform an in vitro trial and faecal bacteria count. In the first evaluation, the faecal inoculum obtained from dogs fed a diet supplemented with hemp showed higher fermentation activity and lower gas production at 24 h of incubation. The bacterial count demonstrated an increase in Lactobacillus when hemp group faeces were tested. Both in vivo and in vitro acetic acid production increased. The results obtained suggest an influence of the fatty acid profile on the microbial population. Abstract Lipids represent a significant energy source in dogs’ diets. Moreover, dogs need some essential fatty acids, such as linoleic and α-linolenic fatty acids, because they are not able to produce them endogenously. This study aimed to evaluate the effect of different dietary lipid sources on faecal microbial populations and activities using different evaluations. Hemp seed oil and swine tallow were tested as lipid supplements in a commercial canned diet at a ratio of 3.5% (HL1 and HL2, respectively). These diets were compared with one rich in starch (HS). Twelve dogs were recruited and equally divided into three groups. Faeces samples at 30 days were used as inoculum and incubated with three different substrates (MOS, inulin, and cellulose) using the in vitro gas production technique. The faecal cell numbers of relevant bacteria and secondary metabolites were analysed (in vivo trial). In vitro evaluation showed that the faeces of the group fed the diet with hemp supplementation had better fermentability despite lower gas production. The in vivo faecal bacterial count showed an increase in Lactobacillus spp. In the HL1 group. Moreover, a higher level of acetate was observed in both evaluations (in vitro and in vivo). These results seem to indicate a significant effect of the dietary fatty acid profile on the faecal microbial population.
Collapse
|
34
|
Jones KA, Richard AJ, Salbaum JM, Newman S, Carmouche R, Webb S, Bruce-Keller AJ, Stephens JM, Campagna SR. Cross-Omics Analysis of Fenugreek Supplementation Reveals Beneficial Effects Are Caused by Gut Microbiome Changes Not Mammalian Host Physiology. Int J Mol Sci 2022; 23:3654. [PMID: 35409014 PMCID: PMC8998956 DOI: 10.3390/ijms23073654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022] Open
Abstract
Herbal remedies are increasing in popularity as treatments for metabolic conditions such as obesity and Type 2 Diabetes. One potential therapeutic option is fenugreek seeds (Trigonella foenum-graecum), which have been used for treating high cholesterol and Type 2 diabetes. A proposed mechanism for these benefits is through alterations in the microbiome, which impact mammalian host metabolic function. This study used untargeted metabolomics to investigate the fenugreek-induced alterations in the intestinal, liver, and serum profiles of mice fed either a 60% high-fat or low-fat control diet each with or without fenugreek supplementation (2% w/w) for 14 weeks. Metagenomic analyses of intestinal contents found significant alterations in the relative composition of the gut microbiome resulting from fenugreek supplementation. Specifically, Verrucomicrobia, a phylum containing beneficial bacteria which are correlated with health benefits, increased in relative abundance with fenugreek. Metabolomics partial least squares discriminant analysis revealed substantial fenugreek-induced changes in the large intestines. However, it was observed that while the magnitude of changes was less, significant modifications were present in the liver tissues resulting from fenugreek supplementation. Further analyses revealed metabolic processes affected by fenugreek and showed broad ranging impacts in multiple pathways, including carnitine biosynthesis, cholesterol and bile acid metabolism, and arginine biosynthesis. These pathways may play important roles in the beneficial effects of fenugreek.
Collapse
Affiliation(s)
- Katarina A. Jones
- Department of Chemistry, University of Tennessee, Knoxville, TN 37916, USA;
| | - Allison J. Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - J. Michael Salbaum
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Susan Newman
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Richard Carmouche
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Sara Webb
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Annadora J. Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA; (A.J.R.); (J.M.S.); (S.N.); (R.C.); (S.W.); (A.J.B.-K.); (J.M.S.)
| | - Shawn R. Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37916, USA;
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN 37916, USA
| |
Collapse
|
35
|
Meliț LE, Mărginean CO, Săsăran MO. The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota. Biomedicines 2022; 10:biomedicines10030645. [PMID: 35327446 PMCID: PMC8945275 DOI: 10.3390/biomedicines10030645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/04/2023] Open
Abstract
The era of pediatric obesity is no longer a myth. Unfortunately, pediatric obesity has reached alarming incidence levels worldwide and the factors that contribute to its development have been intensely studied in multiple recent and emerging studies. Gut microbiota was recently included in the wide spectrum of factors implicated in the determination of obesity, but its role in pediatric obese patients is far from being fully understood. In terms of the infant gut microbiome, multiple factors have been demonstrated to shape its content, including maternal diet and health, type of delivery, feeding patterns, weaning and dietary habits. Nevertheless, the role of the intrauterine environment, such as the placental microbial community, cannot be completely excluded. Most studies have identified Firmicutes and Bacteroidetes as the most important players related to obesity risk in gut microbiota reflecting an increase of Firmicutes and a decrease in Bacteroidetes in the context of obesity; however, multiple inconsistencies between studies were recently reported, especially in pediatric populations, and there is a scarcity of studies performed in this age group.
Collapse
Affiliation(s)
- Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
- Correspondence:
| | - Maria Oana Săsăran
- Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
36
|
Pasupuleti RR, Tsai PC, Ponnusamy VK, Chen NC. Green sample pre-treatment technique coupled with UHPLC-MS/MS for the rapid biomonitoring of dietary poly-unsaturated (omega) fatty acids to predict health risks. CHEMOSPHERE 2022; 291:132685. [PMID: 34743796 DOI: 10.1016/j.chemosphere.2021.132685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/05/2021] [Accepted: 10/23/2021] [Indexed: 06/13/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) consumption indicates beneficial effects on cardiovascular disease (CVD) and physiological processes in humans. However, the inappropriate ratio of omega-(ω)-PUFA levels in human blood is considered as raising the risk of CVD. Therefore, monitoring dietary ω-FAs in human serum is vital for early diagnosis for individuals to predict CVD risk. This work reports a fast green sample pre-treatment protocol for sensitive and simultaneous monitoring of ω-3-FAs and ω-6-FAs in serum by novel in-syringe-based ultrasonication-assisted alkaline hydrolysis coupled with vortex-induced liquid-liquid microextraction (IS-USAH-VI-LLME) technique connected with UHPLC-MS/MS analysis. Factors affecting extraction recoveries of ten ω-PUFAs by the presented method were well-studied. ω-3 and ω-6 PUFAs demonstrated excellent linearities between the concentrations between 0.1-10,000 ng mL-1 with good regression coefficients between 0.9910-0.9997. The detection and quantification limits were between 0.05-0.35 and 0.16-1.07 ng mL-1, demonstrating that the presented method is highly sensitive and versatile. The precision of the technique was <8.2% that deemed acceptable in clinical analysis. Further, the proposed method was applied for ω-PUFAs analysis in human blood samples, and spiked recoveries showed between 80.32-119.34% with <9.82% precision. Results proved that the developed method is green, sensitive, and reliable to simultaneously determine ten ω-PUFAs in human blood samples for clinical diagnosis applications for predicting health hazards.
Collapse
Affiliation(s)
- Raghavendra Rao Pasupuleti
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Pei-Chien Tsai
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, 807, Taiwan; Department of Chemistry, National Sun Yat-sen University (NSYSU), Kaohsiung City, 804, Taiwan; Program of Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, 811, Taiwan.
| | - Nai-Ching Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, 833, Taiwan.
| |
Collapse
|
37
|
Zapata J, Gallardo A, Romero C, Valenzuela R, Garcia-Diaz DF, Duarte L, Bustamante A, Gasaly N, Gotteland M, Echeverria F. n-3 polyunsaturated fatty acids in the regulation of adipose tissue browning and thermogenesis in obesity: Potential relationship with gut microbiota. Prostaglandins Leukot Essent Fatty Acids 2022; 177:102388. [PMID: 34995899 DOI: 10.1016/j.plefa.2021.102388] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/03/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Obesity is a worldwide public health problem characterized by fat tissue accumulation, favouring adipose tissue and metabolic alterations. Increasing energy expenditure (EE) through brown adipose tissue activation and white adipose tissue (WAT) browning has gained relevance as a therapeutic approach. Different bioactive compounds, such as n-3 polyunsaturated fatty acids (PUFA), have been shown to induce those thermogenic effects. This process is regulated by the gut microbiota as well. Nevertheless, obesity is characterized by gut microbiota dysbiosis, which can be restored by weight loss and n-3 PUFA intake, among other factors. Knowledge gap: However, the role of the gut microbiota on the n-3 PUFA effect in inducing thermogenesis in obesity has not been fully elucidated. OBJECTIVE This review aims to elucidate the potential implications of this interrelation on WAT browning adiposw sittue (BAT), BAT activity, and EE regulation in obesity models.
Collapse
Affiliation(s)
- J Zapata
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - A Gallardo
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - C Romero
- Escuela de Medicina, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - R Valenzuela
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Nutritional Sciences Department, Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - D F Garcia-Diaz
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - L Duarte
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - A Bustamante
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - N Gasaly
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; ICBM: Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Chile
| | - M Gotteland
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - F Echeverria
- Departamento de Nutricion, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Carrera de Nutricion y Dietetica, Departamento Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
38
|
Riedel S, Pheiffer C, Johnson R, Louw J, Muller CJF. Intestinal Barrier Function and Immune Homeostasis Are Missing Links in Obesity and Type 2 Diabetes Development. Front Endocrinol (Lausanne) 2022; 12:833544. [PMID: 35145486 PMCID: PMC8821109 DOI: 10.3389/fendo.2021.833544] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Noncommunicable diseases, such as type 2 diabetes (T2D), place a burden on healthcare systems worldwide. The rising prevalence of obesity, a major risk factor for T2D, is mainly attributed to the adoption of Westernized diets and lifestyle, which cause metabolic dysfunction and insulin resistance. Moreover, diet may also induce changes in the microbiota composition, thereby affecting intestinal immunity. The critical role of intestinal immunity and intestinal barrier function in the development of T2D is increasingly acknowledged, however, limited studies have investigated the link between intestinal function and metabolic disease. In this review, studies reporting specific roles of the intestinal immune system and intestinal epithelial cells (IECs) in metabolic disease are highlighted. Innate chemokine signaling, eosinophils, immunoglobulin A (IgA), T helper (Th) 17 cells and their cytokines were associated with obesity and/or dysregulated glucose homeostasis. Intestinal epithelial cells (IECs) emerged as critical modulators of obesity and glucose homeostasis through their effect on lipopolysaccharide (LPS) signaling and decontamination. Furthermore, IECs create a link between microbial metabolites and whole-body metabolic function. Future in depth studies of the intestinal immune system and IECs may provide new opportunities and targets to develop treatments and prevention strategies for obesity and T2D.
Collapse
Affiliation(s)
- Sylvia Riedel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| |
Collapse
|
39
|
Choi SI, Kim N, Nam RH, Park JH, Nho H, Yu JE, Song CH, Lee SM, Lee DH. Fecal Microbial Enterotypes Differentially Respond to a High-fat Diet Based on Sex in Fischer-344 Rats. J Cancer Prev 2021; 26:277-288. [PMID: 35047454 PMCID: PMC8749319 DOI: 10.15430/jcp.2021.26.4.277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 11/03/2022] Open
Abstract
The gut microbiota interacts with the host gut environment, which is influenced by such factors as sex, age, and host diet. These factors induce changes in the microbial composition. The aim of this study was to identify differences in the gut microbiome of Fisher-344 (F344) rats fed a high-fat diet (HFD), depending on their age and sex. Fecal microbiomes from 6-, 31-, and 74-week-old, and 2-year-old both male and female rats (corresponding to 5-, 30-, 60-, and 80-year-old humans) were analyzed using 16S rRNA gene sequencing, phylogenetic investigation of communities by reconstruction of unobserved states, and enterotype (E) assessment. Moreover, the effect of an HFD on colonic epithelial cells was measured using real-time quantitative PCR. Alpha diversity decreased in the HFD group regardless of age and sex. Based on the enterotype clustering of the whole fecal microbiome, clusters from male rats were divided into E1 and E2 enterotypes, while clusters from female rats were divided into E1, E2, and E3 enterotypes. The female E3 group showed a significantly high abundance in the Ruminococcus genus and expression of Tlr2 mRNA, which may reflect compensation to the HFD. Moreover, the female E3 group showed a lower ratio of opportunistic pathogenic strains to commensal strains compared to the female E2 group. Administration of an HFD influenced the rat fecal microbiota in all assessed age groups, which could be further differentiated by sex. In particular, female rats showed a compensatory enterotype response to an HFD compared to male rats.
Collapse
Affiliation(s)
- Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Heewon Nho
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jeong Eun Yu
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sun Min Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Mikrobiota jelitowa a leki. Interakcje wpływające na skuteczność i bezpieczeństwo farmakoterapii. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
Mikrobiota jelitowa stanowi nieodłączny element organizmu umożliwiający jego prawidłowe funkcjonowanie. Dzięki mikroorganizmom jelitowym możliwa jest stymulacja układu odpornościowego, synteza witamin czy poprawa wchłaniania składników odżywczych. Jednak jej aktywność może również niekorzystnie działać na organizm, m.in. z powodu przetwarzania treści jelitowej. Opisywana w artykule interakcja mikrobiota–lek uwzględnia pozytywny i negatywny wpływ mikroorganizmów jelitowych na farmakoterapię poprzez bezpośrednie i pośrednie oddziaływanie na lek w organizmie. Ze względu na to, że mikrobiom stanowi nieodłączny element organizmu, ingerencja nawet w jego niewielką część może doprowadzić do wystąpienia daleko idących, czasami niespodziewanych skutków. Stąd w celu poprawy skuteczności i bezpieczeństwa farmakoterapii konieczne jest wyjaśnienie mechanizmów oddziaływania mikrobioty na lek w organizmie.
W artykule podsumowano obecną wiedzę na temat biologicznej aktywności mikrobioty jelitowej, a zwłaszcza oddziaływań mikrobiota–leki determinujących skuteczność i bezpieczeństwo farmakoterapii. Wyszukiwanie przeprowadzono we wrześniu 2020 r. w bazach danych PubMed, Scopus, Web of Science, Cochrane Library i powszechnie dostępnej literaturze z użyciem terminów: „mikrobiota jelitowa”, „mikrobiom”, „metabolizm leku”, „interakcje mikrobiota–lek”. W artykule omówiono interakcje między mikrobiotą a lekami m.in. z grupy antybiotyków, inhibitorów pompy protonowej, sulfonamidów, pochodnych kwasu 5-aminosalicylowego, niesteroidowych leków przeciwzapalnych, przeciwnowotworowych, statyn czy metforminą.
Collapse
|
41
|
Peters V, Bolte L, Schuttert E[M, Andreu-Sánchez S, Dijkstra G, Weersma R[K, Campmans-Kuijpers M[JE. Western and Carnivorous Dietary Patterns are Associated with Greater Likelihood of IBD Development in a Large Prospective Population-based Cohort. J Crohns Colitis 2021; 16:931-939. [PMID: 34864946 PMCID: PMC9282880 DOI: 10.1093/ecco-jcc/jjab219] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/09/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Nutrition plays a role in the development of Crohn's disease [CD] and ulcerative colitis [UC]. However, prospective data on nutrition and disease onset are limited. Here, we analysed dietary patterns and scores in relation to inflammatory bowel disease [IBD] development in a prospective population-based cohort. METHODS We analysed 125 445 participants of whom 224 individuals developed de novo UC and 97 CD over a maximum 14-year follow-up period. Participants answered health-related [also prospectively] and dietary questionnaires [FFQ] at baseline. Principal component analysis [PCA] was conducted deriving a-posteriori dietary patterns. Hypotheses-based a-priori dietary scores were also calculated, including the protein score, Healthy Eating Index, LifeLines Diet Score [LLDS], and alternative Mediterranean Diet Score. Logistic regression models were performed between dietary patterns, scores, and IBD development. RESULTS PCA identified five dietary patterns. A pattern characterised by high intake of snacks, prepared meals, non-alcoholic beverages, and sauces along with low vegetables and fruit consumption was associated with higher likelihood of CD development (odds ratio [OR]: 1.16, 95% confidence interval [CI]: 1.03-1.30, p = 0.013). A pattern comprising red meat, poultry, and processed meat, was associated with increased likelihood of UC development [OR: 1.11, 95% CI: 1.01-1.20, p = 0.023]. A high diet quality score [LLDS] was associated with decreased risk of CD [OR: 0.95, 95% CI: 0.92-0.99, p = 0.009]. CONCLUSIONS A Western dietary pattern was associated with a greater likelihood of CD development and a carnivorous pattern with UC development, whereas a relatively high diet quality [LLDS] was protective for CD development. Our study strengthens the importance of evaluating dietary patterns to aid prevention of IBD in the general population.
Collapse
Affiliation(s)
- Vera Peters
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, The Netherlands,Department of Epidemiology, University of Groningen and University Medical Centre Groningen, The Netherlands
| | - Laura Bolte
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, The Netherlands,Department of Genetics, University of Groningen and University Medical Centre Groningen, The Netherlands
| | - Eva [Monique] Schuttert
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, The Netherlands,Department of Genetics, University of Groningen and University Medical Centre Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Centre Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, The Netherlands
| | | | - Marjo [Johanna Elisabeth] Campmans-Kuijpers
- Corresponding author: Marjo [Johanna Elisabeth] Campmans-Kuijpers, University Medical Centre Groningen, Hanzeplein 1, PO-box 30.001, 9700 RB, Department of Gastroenterology, HPC BB41, Groningen, The Netherlands. Tel.: +31 [0]50 361 2620; fax: +31 [0]50 361 9306;
| |
Collapse
|
42
|
Hughes RL, Holscher HD. Fueling Gut Microbes: A Review of the Interaction between Diet, Exercise, and the Gut Microbiota in Athletes. Adv Nutr 2021; 12:2190-2215. [PMID: 34229348 PMCID: PMC8634498 DOI: 10.1093/advances/nmab077] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/19/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The athlete's goal is to optimize their performance. Towards this end, nutrition has been used to improve the health of athletes' brains, bones, muscles, and cardiovascular system. However, recent research suggests that the gut and its resident microbiota may also play a role in athlete health and performance. Therefore, athletes should consider dietary strategies in the context of their potential effects on the gut microbiota, including the impact of sports-centric dietary strategies (e.g., protein supplements, carbohydrate loading) on the gut microbiota as well as the effects of gut-centric dietary strategies (e.g., probiotics, prebiotics) on performance. This review provides an overview of the interaction between diet, exercise, and the gut microbiota, focusing on dietary strategies that may impact both the gut microbiota and athletic performance. Current evidence suggests that the gut microbiota could, in theory, contribute to the effects of dietary intake on athletic performance by influencing microbial metabolite production, gastrointestinal physiology, and immune modulation. Common dietary strategies such as high protein and simple carbohydrate intake, low fiber intake, and food avoidance may adversely impact the gut microbiota and, in turn, performance. Conversely, intake of adequate dietary fiber, a variety of protein sources, and emphasis on unsaturated fats, especially omega-3 (ɷ-3) fatty acids, in addition to consumption of prebiotics, probiotics, and synbiotics, have shown promising results in optimizing athlete health and performance. Ultimately, while this is an emerging and promising area of research, more studies are needed that incorporate, control, and manipulate all 3 of these elements (i.e., diet, exercise, and gut microbiome) to provide recommendations for athletes on how to "fuel their microbes."
Collapse
Affiliation(s)
- Riley L Hughes
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Nutrition Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
43
|
Jiang Y, Rodgers B, Damiris K, Choi C, Ahlawat S. The effects of diabetes mellitus on clinical outcomes of hospitalized patients with acute diverticulitis. Eur J Gastroenterol Hepatol 2021; 33:1354-1360. [PMID: 32796358 DOI: 10.1097/meg.0000000000001895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Acute diverticulitis is a common gastrointestinal illness due to diverticular inflammation and focal necrosis. Diabetes mellitus has been reported to influence the outcomes of patients with diverticular disease. Our study aimed to examine the inpatient outcomes and complications of patients with acute diverticulitis and coexisting diabetes mellitus. METHODS The Nationwide Inpatient Sample was used to identify adult patients in 2014 admitted for acute diverticulitis. Primary outcomes were mortality, length of stay (LOS), and total hospitalization charges. Secondary outcomes were complications of acute diverticulitis and interventions. RESULTS In total, 44 330 of patients with acute diverticulitis and diabetes mellitus were included in the analysis. Acute diverticulitis patients with diabetes mellitus had a higher rate of diverticular bleeding (P < 0.0001), but lower rates of abscess (P < 0.0001), obstruction (P < 0.0001) and colectomy (P < 0.0001) when compared to acute diverticulitis patients without diabetes mellitus. Complicated diabetes mellitus was associated with a longer LOS (P = 0.00003) and greater total hospitalization charges (P = 0.0021) compared to uncomplicated diabetes mellitus when coexisting with acute diverticulitis. CONCLUSIONS Acute diverticulitis with diabetes mellitus is associated with a higher rate of diverticular bleeding, lower rates of abscess, obstruction, and colectomy compared to acute diverticulitis without diabetes mellitus. When coexisting with acute diverticulitis, complicated diabetes mellitus is not associated with higher rates of mortality or diverticulitis-related complications compared to uncomplicated diabetes mellitus.
Collapse
Affiliation(s)
| | | | | | | | - Sushil Ahlawat
- Division of Gastroenterology and Hepatology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
44
|
Zubareva OE, Melik-Kasumov TB. The Gut–Brain Axis and Peroxisome Proliferator-Activated Receptors in the Regulation of Epileptogenesis. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Zhong H, Abdullah, Deng L, Zhao M, Tang J, Liu T, Zhang H, Feng F. Probiotic-fermented blueberry juice prevents obesity and hyperglycemia in high fat diet-fed mice in association with modulating the gut microbiota. Food Funct 2021; 11:9192-9207. [PMID: 33030465 DOI: 10.1039/d0fo00334d] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Blueberry dietary interventions have demonstrated remarkable potential against obesity and type 2 diabetes mellitus. However, the effects of fermented blueberry juice on metabolic syndrome, the gut microbiota, and insulin resistance have not yet been reported. This study aimed to investigate the potential of fermented blueberry juice against obesity, hyperglycemia, and gut microbiota dysbiosis in high fat diet (HFD)-fed mice. Our study findings revealed that supplementation with fresh blueberry juice (BBJ), and fermented blueberry juice with homemade probiotic starter (FBJ) or commercial starter (CFBJ) significantly decreased fat accumulation and low density lipoprotein cholesterol (LDL-C) levels in HFD-fed mice. FBJ showed relatively more potency to reduce body weight than BBJ and CFBJ. The percentage increase in the body weight of the FBJ group was almost the same as that in the normal chow diet (NCD) group, and was approximately 10% lower than the BBJ and CFBJ groups. Overall, all blueberry juices significantly ameliorated hyperlipidemia and insulin resistance in HFD-fed mice. Moreover, the dietary interventions with BBJ, FBJ, and CFBJ for 17 weeks significantly improved the community richness and diversity of the gut microflora along with an altered structure in the HFD-fed mice group. The FBJ treated mice group showed relatively low abundance of Firmicutes, obesity-related bacteria (Oscillibacter and Alistipes), and high abundance of lean bacteria (Akkermansia, Barnesiella, Olsenella, Bifidobacterium, and Lactobacillus) compared to the HFD-fed mice group. Furthermore, BBJ and FBJ treatments regulated the liver mRNA and protein expression levels involved in lipid and glucose metabolism. This study inferred that fermented blueberry juice could be used as a functional food to prevent the modern pandemics i.e., obesity and insulin resistance.
Collapse
Affiliation(s)
- Hao Zhong
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| | - Abdullah
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China.
| | - Lingli Deng
- College of Biological Science and Technology, Hubei Minzu University, Enshi 445000, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| | - Jun Tang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| | - Tao Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| | - Hui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China. and Ningbo Institute of Zhejiang University, Ningbo 315100, China
| |
Collapse
|
46
|
Bolte LA, Vich Vila A, Imhann F, Collij V, Gacesa R, Peters V, Wijmenga C, Kurilshikov A, Campmans-Kuijpers MJE, Fu J, Dijkstra G, Zhernakova A, Weersma RK. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut 2021; 70:1287-1298. [PMID: 33811041 PMCID: PMC8223641 DOI: 10.1136/gutjnl-2020-322670] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The microbiome directly affects the balance of pro-inflammatory and anti-inflammatory responses in the gut. As microbes thrive on dietary substrates, the question arises whether we can nourish an anti-inflammatory gut ecosystem. We aim to unravel interactions between diet, gut microbiota and their functional ability to induce intestinal inflammation. DESIGN We investigated the relation between 173 dietary factors and the microbiome of 1425 individuals spanning four cohorts: Crohn's disease, ulcerative colitis, irritable bowel syndrome and the general population. Shotgun metagenomic sequencing was performed to profile gut microbial composition and function. Dietary intake was assessed through food frequency questionnaires. We performed unsupervised clustering to identify dietary patterns and microbial clusters. Associations between diet and microbial features were explored per cohort, followed by a meta-analysis and heterogeneity estimation. RESULTS We identified 38 associations between dietary patterns and microbial clusters. Moreover, 61 individual foods and nutrients were associated with 61 species and 249 metabolic pathways in the meta-analysis across healthy individuals and patients with IBS, Crohn's disease and UC (false discovery rate<0.05). Processed foods and animal-derived foods were consistently associated with higher abundances of Firmicutes, Ruminococcus species of the Blautia genus and endotoxin synthesis pathways. The opposite was found for plant foods and fish, which were positively associated with short-chain fatty acid-producing commensals and pathways of nutrient metabolism. CONCLUSION We identified dietary patterns that consistently correlate with groups of bacteria with shared functional roles in both, health and disease. Moreover, specific foods and nutrients were associated with species known to infer mucosal protection and anti-inflammatory effects. We propose microbial mechanisms through which the diet affects inflammatory responses in the gut as a rationale for future intervention studies.
Collapse
Affiliation(s)
- Laura A Bolte
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Floris Imhann
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Valerie Collij
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Ranko Gacesa
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Vera Peters
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Marjo J E Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
47
|
Olvera-Rosales LB, Cruz-Guerrero AE, Ramírez-Moreno E, Quintero-Lira A, Contreras-López E, Jaimez-Ordaz J, Castañeda-Ovando A, Añorve-Morga J, Calderón-Ramos ZG, Arias-Rico J, González-Olivares LG. Impact of the Gut Microbiota Balance on the Health-Disease Relationship: The Importance of Consuming Probiotics and Prebiotics. Foods 2021; 10:1261. [PMID: 34199351 PMCID: PMC8230287 DOI: 10.3390/foods10061261] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota is a group of microorganisms that are deposited throughout the entire gastrointestinal tract. Currently, thanks to genomic tools, studies of gut microbiota have pointed towards the understanding of the metabolism of important bacteria that are not cultivable and their relationship with human homeostasis. Alterations in the composition of gut microbiota could explain, at least in part, some epidemics, such as diabetes and obesity. Likewise, dysbiosis has been associated with gastrointestinal disorders, neurodegenerative diseases, and even cancer. That is why several studies have recently been focused on the direct relationship that these types of conditions have with the specific composition of gut microbiota, as in the case of the microbiota-intestine-brain axis. In the same way, the control of microbiota is related to the diet. Therefore, this review highlights the importance of gut microbiota, from its composition to its relationship with the human health-disease condition, as well as emphasizes the effect of probiotic and prebiotic consumption on the balance of its composition.
Collapse
Affiliation(s)
- Laura-Berenice Olvera-Rosales
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| | - Alma-Elizabeth Cruz-Guerrero
- Departamento de Biotecnología, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Av. San Rafael Atlixco 186, Ciudad de Mexico 09340, Mexico
| | - Esther Ramírez-Moreno
- Área Académica de Nutrición, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda, La Concepción S/N, Carretera Pachuca Actopan, San Agustín Tlaxiaca 42060, Hidalgo, Mexico; (E.R.-M.); (Z.-G.C.-R.)
| | - Aurora Quintero-Lira
- Área Académica de Ingeniería Agroindustrial e Ingeniería en alimentos, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km. 1, Ex-Hacienda de Aquetzalpa, Tulancingo 43600, Hidalgo, Mexico;
| | - Elizabeth Contreras-López
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| | - Judith Jaimez-Ordaz
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| | - Araceli Castañeda-Ovando
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| | - Javier Añorve-Morga
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| | - Zuli-Guadalupe Calderón-Ramos
- Área Académica de Nutrición, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda, La Concepción S/N, Carretera Pachuca Actopan, San Agustín Tlaxiaca 42060, Hidalgo, Mexico; (E.R.-M.); (Z.-G.C.-R.)
| | - José Arias-Rico
- Área Académica de Enfermería, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda, La Concepción S/N, Carretera Pachuca Actopan, San Agustín Tlaxiaca 42060, Hidalgo, Mexico;
| | - Luis-Guillermo González-Olivares
- Área Académica de Química, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma 42184, Hidalgo, Mexico; (L.-B.O.-R.); (E.C.-L.); (J.J.-O.); (A.C.-O.); (J.A.-M.)
| |
Collapse
|
48
|
Han C, Guo N, Bu Y, Peng Y, Li X, Ma X, Yang M, Jia X, Zhang J, Liu X, Yu K, Wang C. Intestinal microbiota and antibiotic-associated acute gastrointestinal injury in sepsis mice. Aging (Albany NY) 2021; 13:10099-10111. [PMID: 33818419 PMCID: PMC8064167 DOI: 10.18632/aging.202768] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/18/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND To investigate the changes of intestinal microbiota and metabolites in sepsis mice with acute gastrointestinal injury before and after the use of antibiotics, and to explore the possible effects of these changes on the body. METHODS Twenty-four 6-8-w-old SPF-grade C57BL/6J male mice were selected, and the mice were randomly divided into three groups. The mice were treated by tail vein injection for 3 days. The intestinal motility of mice after administration was detected. The mice feces were collected for 16S rRNA and Untargeted metabonomics detection. RESULTS The use of antibiotics in sepsis mice can change the composition of intestinal microbiota and metabolites. LD3, AD3 and LAD3 samples had significant differences in bacterial species. Desulfovibrio was the species with a significant difference in LAD3. In addition, we found that the composition of those intestinal microbiota were correlated with changes in intestinal motility. The untargeted metabolomics analysis showed that the fecal metabolites of LD3 and LAD3 samples were significantly different. In addition to the basic metabolites, Benzoic acid and 4-Hydroxybenzoic acid were also found, and Desulfovibrio was associated with them. CONCLUSIONS The use of antibiotics in sepsis mice can lead to changes in the intestinal microbiota and metabolite levels, which may be related to the severity of acute gastrointestinal injury in sepsis mice. Inhibiting Desulfovibrio in the intestine and using Benzoic acid and 4-Hydroxybenzoic acid as a marker for the production of Desulfovibrio may reduce the inflammatory degree of acute gastrointestinal injury in sepsis.
Collapse
Affiliation(s)
- Ci Han
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Nana Guo
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yue Bu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yahui Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xueting Li
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Xiaohui Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Mengyuan Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiaonan Jia
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Jin Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiaowei Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Changsong Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin 150001, Heilongjiang, China.,Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang, China
| |
Collapse
|
49
|
Ji XW, Wang J, Shen QM, Li ZY, Jiang YF, Liu DK, Tan YT, Li HL, Xiang YB. Dietary fat intake and liver cancer incidence: A population-based cohort study in Chinese men. Int J Cancer 2021; 148:2982-2996. [PMID: 33559177 DOI: 10.1002/ijc.33507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
To date, limited studies have focused on the association between dietary fat and liver cancer risk, especially in China. Our study aims to evaluate the association between dietary fat intake and liver cancer incidence risk in men. Dietary fat intake was obtained through a validated food frequency questionnaire in a Chinese prospective cohort. The Cox regression model was utilized to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). After exclusion, 59 998 recruitments were finally analyzed with a total follow-up time of 714 339 person-years, 431 incident liver cancer cases were newly identified among them. The adjusted HRs (95% CIs) for the highest vs lowest quartile of total fat, saturated fat, monounsaturated fat (MUFA), and polyunsaturated fat (PUFA) were 1.33 (1.01-1.75), 1.50 (1.13-1.97), 1.26 (0.96-1.65), and 1.41 (1.07-1.86), and the corresponding P-trend values were .008, .005, .034, and .005, respectively. In the secondary analysis among participants tested for hepatitis B virus, we found that higher intakes of saturated fat and PUFA were also associated with increased liver cancer risks. Besides, high risks of per standard deviation alterations of the total fat, saturated fat and MUFA were detected in liver cancer, and these results were similar to those concluded from the full-cohort analysis. In conclusion, dietary intakes of total fat, saturated fat, PUFA, and probably MUFA might increase liver cancer risks. Our study provides suggestive advice to public administration on dietary suggestions, and related measures taken from managing dietary fat intake might reduce liver cancer incidence.
Collapse
Affiliation(s)
- Xiao-Wei Ji
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jing Wang
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qiu-Ming Shen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhuo-Ying Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yu-Fei Jiang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Da-Ke Liu
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yu-Ting Tan
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hong-Lan Li
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yong-Bing Xiang
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
50
|
Mehl C, Schoeman MC, Sanko TJ, Bezuidenhout C, Mienie CMS, Preiser W, Vosloo D. Wastewater treatment works change the intestinal microbiomes of insectivorous bats. PLoS One 2021; 16:e0247475. [PMID: 33657147 PMCID: PMC7928523 DOI: 10.1371/journal.pone.0247475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Mammals, born with a near-sterile intestinal tract, are inoculated with their mothers’ microbiome during birth. Thereafter, extrinsic and intrinsic factors shape their intestinal microbe assemblage. Wastewater treatment works (WWTW), sites synonymous with pollutants and pathogens, receive influent from domestic, agricultural and industrial sources. The high nutrient content of wastewater supports abundant populations of chironomid midges (Diptera), which transfer these toxicants and potential pathogens to their predators, such as the banana bat Neoromicia nana (Vespertilionidae), thereby influencing their intestinal microbial assemblages. We used next generation sequencing and 16S rRNA gene profiling to identify and compare intestinal bacteria of N. nana at two reference sites and two WWTW sites. We describe the shared intestinal microbiome of the insectivorous bat, N. nana, consisting of seven phyla and eleven classes. Further, multivariate analyses revealed that location was the most significant driver (sex, body size and condition were not significant) of intestinal microbiome diversity. Bats at WWTW sites exhibited greater intestinal microbiota diversity than those at reference sites, likely due to wastewater exposure, stress and/or altered diet. Changes in their intestinal microbiota assemblages may allow these bats to cope with concomitant stressors.
Collapse
Affiliation(s)
- Calvin Mehl
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - M. Corrie Schoeman
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Tomasz J. Sanko
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Carlos Bezuidenhout
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Charlotte M. S. Mienie
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Wolfgang Preiser
- Division of Medical Virology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, South Africa
- National Health Laboratory Service (NHLS), Tygerberg Hospital, Tygerberg, South Africa
| | - Dalene Vosloo
- Centre for Functional Biodiversity, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
- * E-mail:
| |
Collapse
|