1
|
Role of a mixed probiotic product, VSL#3, in the prevention and treatment of colorectal cancer. Eur J Pharmacol 2022; 930:175152. [PMID: 35835181 DOI: 10.1016/j.ejphar.2022.175152] [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: 05/19/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 12/09/2022]
Abstract
Colorectal cancer (CRC) is a multifactorial disease. The incidence of this type of cancer in younger patients has increased in recent years, and more strategies are needed to prevent and delay the progression of CRC. Probiotics play an adjunctive role in the prevention and treatment of CRC and can not only prevent the onset and delay the progression of disease but also reduce the side effects after the application of anti-cancer drugs. The anti-cancer effect of individual probiotics has been extensively studied, and the exact curative effect of various probiotics has been found, but the anti-cancer effect of mixed probiotics is still not well summarized. In this review, we discuss the positive effects of mixed probiotics on CRC and the related mechanisms of action, especially VSL#3 (VSL Pharmaceuticals, Inc., Gaithersburg, MD, USA), thus providing new ideas for the treatment of CRC. Moreover, we suggest the need to search for more therapeutic possibilities, especially via the research and application of synbiotics and postbiotics.
Collapse
|
2
|
Wang X, Zhao J, Feng Y, Feng Z, Ye Y, Liu L, Kang G, Cao X. Evolutionary Insights Into Microbiota Transplantation in Inflammatory Bowel Disease. Front Cell Infect Microbiol 2022; 12:916543. [PMID: 35811664 PMCID: PMC9257068 DOI: 10.3389/fcimb.2022.916543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The intestinal microbiome plays an essential role in human health and disease status. So far, microbiota transplantation is considered a potential therapeutic approach for treating some chronic diseases, including inflammatory bowel disease (IBD). The diversity of gut microbiota is critical for maintaining resilience, and therefore, transplantation with numerous genetically diverse gut microbiota with metabolic flexibility and functional redundancy can effectively improve gut health than a single probiotic strain supplement. Studies have shown that natural fecal microbiota transplantation or washing microbiota transplantation can alleviate colitis and improve intestinal dysbiosis in IBD patients. However, unexpected adverse reactions caused by the complex and unclear composition of the flora limit its wider application. The evolving strain isolation technology and modifiable pre-existing strains are driving the development of microbiota transplantation. This review summarized the updating clinical and preclinical data of IBD treatments from fecal microbiota transplantation to washing microbiota transplantation, and then to artificial consortium transplantation. In addition, the factors considered for strain combination were reviewed. Furthermore, four types of artificial consortium transplant products were collected to analyze their combination and possible compatibility principles. The perspective on individualized microbiota transplantation was also discussed ultimately.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yuanhang Feng
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Zelin Feng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yulin Ye
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Limin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Guangbo Kang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
- *Correspondence: Xiaocang Cao, ; Guangbo Kang,
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- *Correspondence: Xiaocang Cao, ; Guangbo Kang,
| |
Collapse
|
3
|
Cell-free probiotic supernatant (CFS) treatment alleviates indomethacin-induced enterocolopathy in BALB/c mice by down-modulating inflammatory response and oxidative stress: potential alternative targeted treatment. Inflammopharmacology 2022; 30:1685-1703. [PMID: 35505268 DOI: 10.1007/s10787-022-00996-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/08/2022] [Indexed: 11/05/2022]
Abstract
Probiotics and their metabolites appear to be a promising approach that targets both the intestinal inflammation and dysbiosis in bowel diseases. In this context, the emergence of the probiotic cell-free supernatant (CFS) has attracted more attention as a safe and targeted alternative therapy with reduced side effects. The use of nonsteroidal anti-inflammatory drugs (NSAIDs) can cause significant intestinal alterations and inflammation, leading to experimental enterocolopathy resembling Crohn disease. Therefore, we investigated the effect of CFS supplementation on the inflammation and the mucosal intestinal alterations induced by NSAIDs, indomethacin. In the current study, a murine model of intestinal inflammation was generated by the oral gavage (o.g) of indomethacin (10 mg/kg) to BALB/C mice. A group of mice treated with indomethacin was concomitantly treated orally by CFS for 5 days. The Body Health Condition index was monitored, and histological scores were evaluated. Moreover, oxidative and pro-inflammatory markers were assessed. Interestingly, we observed that CFS treatment attenuated the severity of the intestinal inflammation in our enterocolopathy model and resulted in the improvement of the clinical symptoms and the histopathological features. Notably, nitric oxide, tumor necrosis factor alpha, malondialdehyde, and myeloperoxidase levels were down-modulated by CFS supplementation. Concomitantly, an attenuation of NF-κB p65, iNOS, COX2 expression in the ileum and the colon was reported. Collectively, our data suggest that CFS treatment has a beneficial effect in experimental enterocolopathy model and could constitute a good therapeutic candidate for alleviating inflammatory responses and to maintain mucosal homeostasis during chronic and severe conditions of intestinal inflammation.
Collapse
|
4
|
Guo X, Huang C, Xu J, Xu H, Liu L, Zhao H, Wang J, Huang W, Peng W, Chen Y, Nie Y, Zhou Y, Zhou Y. Gut Microbiota Is a Potential Biomarker in Inflammatory Bowel Disease. Front Nutr 2022; 8:818902. [PMID: 35127797 PMCID: PMC8814525 DOI: 10.3389/fnut.2021.818902] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), is characterized by relapse and remission alternately. It remains a great challenge to diagnose and assess disease activity during IBD due to the lack of specific markers. While traditional biomarkers from plasma and stool, such as C-reactive protein (CRP), fecal calprotectin (FC), and S100A12, can be used to measure inflammation, they are not specific to IBD and difficult to determine an effective cut-off value. There is consensus that gut microbiota is crucial for intestinal dysbiosis is closely associated with IBD etiopathology and pathogenesis. Multiple studies have documented differences in the composition of gut microbiota between patients with IBD and healthy individuals, particularly regarding microbial diversity and relative abundance of specific bacteria. Patients with IBD have higher levels of Proteobacteria and lower amounts of Bacteroides, Eubacterium, and Faecalibacterium than healthy individuals. This review summarizes the pros and cons of using traditional and microbiota biomarkers to assess disease severity and treatment outcomes and addresses the possibility of using microbiota-focused interventions during IBD treatment. Understanding the role of microbial biomarkers in the assessment of disease activity and treatment outcomes has the potential to change clinical practice and lead to the development of more personalized therapies.
Collapse
Affiliation(s)
- Xue Guo
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Le Liu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Hailan Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiaqi Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wenqi Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wu Peng
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Yongjian Zhou
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Youlian Zhou
| |
Collapse
|
5
|
|
6
|
Abstract
Inflammatory Bowel Disease (IBD) is a term used to describe a group of complex disorders of the gastrointestinal (GI) tract. IBDs include two main forms: Crohn’s Disease (CD) and Ulcerative Colitis (UC), which share similar clinical symptoms but differ in the anatomical distribution of the inflammatory lesions. The etiology of IBDs is undetermined. Several hypotheses suggest that Crohn’s Disease and Ulcerative Colitis result from an abnormal immune response against endogenous flora and luminal antigens in genetically susceptible individuals. While there is no cure for IBDs, most common treatments (medication and surgery) aim to reduce inflammation and help patients to achieve remission. There is growing evidence and focus on the prophylactic and therapeutic potential of probiotics in IBDs. Probiotics are live microorganisms that regulate the mucosal immune system, the gut microbiota and the production of active metabolites such as Short-Chain Fatty Acids (SCFAs). This review will focus on the role of intestinal dysbiosis in the immunopathogenesis of IBDs and understanding the health-promoting effects of probiotics and their metabolites.
Collapse
|
7
|
Cruz BCDS, Conceição LLD, Mendes TADO, Ferreira CLDLF, Gonçalves RV, Peluzio MDCG. Use of the synbiotic VSL#3 and yacon-based concentrate attenuates intestinal damage and reduces the abundance of Candidatus Saccharimonas in a colitis-associated carcinogenesis model. Food Res Int 2020; 137:109721. [PMID: 33233290 DOI: 10.1016/j.foodres.2020.109721] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/22/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Individuals with inflammatory bowel disease are at high risk of developing colitis-associated cancer; thus, strategies to inhibit disease progression should be investigated. The study aimed to explore the role of the synbiotic (probiotic VSL#3® and yacon-based concentrate) in a colitis-associated carcinogenesis model. IL-10-/- mice were induced to carcinogenesis with 1,2-dimethylhydrazine and divided into two experimental groups: control and synbiotic. Manifestations of colitis, colon histology, expression of antioxidant enzymes, production of organic acids and intestinal microbiota were evaluated. The use of the synbiotic showed benefits, such as the preservation of intestinal architecture, increased expression of antioxidant enzymes and the concentration of organic acids, especially butyrate. It was also observed different microbial community profiles between the groups during the study. Together, these factors contributed to mitigate the manifestations of colitis and improve intestinal integrity, suggesting the potential benefit of the synbiotic in intestinal diseases.
Collapse
Affiliation(s)
- Bruna Cristina Dos Santos Cruz
- Nutritional Biochemistry Laboratory, Department of Nutrition and Health, Universidade Federal de Viçosa - UFV, Viçosa, Minas Gerais, Brazil.
| | - Lisiane Lopes da Conceição
- Nutritional Biochemistry Laboratory, Department of Nutrition and Health, Universidade Federal de Viçosa - UFV, Viçosa, Minas Gerais, Brazil.
| | | | | | | | - Maria do Carmo Gouveia Peluzio
- Nutritional Biochemistry Laboratory, Department of Nutrition and Health, Universidade Federal de Viçosa - UFV, Viçosa, Minas Gerais, Brazil.
| |
Collapse
|
8
|
Synbiotic VSL#3 and yacon-based product modulate the intestinal microbiota and prevent the development of pre-neoplastic lesions in a colorectal carcinogenesis model. Appl Microbiol Biotechnol 2020; 104:8837-8857. [DOI: 10.1007/s00253-020-10863-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
|
9
|
Lu R, Shang M, Zhang YG, Jiao Y, Xia Y, Garrett S, Bakke D, Bäuerl C, Martinez GP, Kim CH, Kang SM, Sun J. Lactic Acid Bacteria Isolated From Korean Kimchi Activate the Vitamin D Receptor-autophagy Signaling Pathways. Inflamm Bowel Dis 2020; 26:1199-1211. [PMID: 32170938 PMCID: PMC7365811 DOI: 10.1093/ibd/izaa049] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Probiotic lactic acid bacteria (LAB) have been used in the anti-inflammation and anti-infection process of various diseases, including inflammatory bowel disease (IBD). Vitamin D receptor (VDR) plays an essential role in pathogenesis of IBD and infectious diseases. Previous studies have demonstrated that the human VDR gene is a key host factor to shape gut microbiome. Furthermore, intestinal epithelial VDR conditional knockout (VDRΔIEC) leads to dysbiosis. Low expressions of VDR is associated with impaired autophagy, accompanied by a reduction of ATG16L1 and LC3B. The purpose of this study is to investigate probiotic effects and mechanism in modulating the VDR-autophagy pathways. METHODS Five LAB strains were isolated from Korean kimchi. Conditional medium (CM) from these strains was used to treat a human cell line HCT116 or intestinal organoids to measure the expression of VDR and autophagy. Mouse embryonic fibroblast (MEF) cells with or without VDR were used to investigate the dependence on the VDR signaling. To test the role of LAB in anti-inflammation, VDR+/+ organoids were treated with 121-CM before infection with Salmonella enterica serovar Enteritidis. In vivo, the role of LAB in regulating VDR-autophagy signaling was examined using LAB 121-CM orally administrated to VDRLoxp and VDRΔIEC mice. RESULTS The LAB-CM-treated groups showed higher mRNA expression of VDR and its target genes cathelicidin compared with the control group. LAB treatment also enhanced expressions of Beclin-1 and ATG16L1 and changed the ratio of LC3B I and II, indicating the activation of autophagic responses. Furthermore, 121-CM treatment before Salmonella enterica serovar Enteritidis infection dramatically increased VDR and ATG16L1 and inhibited the inflammation. Administration of 121-CM to VDRLoxp and VDRΔIEC mice for 12 and 24 hours resulted in an increase of VDR and LC3B II:I ratio. Furthermore, we identified that probiotic proteins P40 and P75 in the LAB-CM contributed to the anti-inflammatory function by increasing VDR. CONCLUSIONS Probiotic LAB exert anti-inflammation activity and induces autophagy. These effects depend on the VDR expression. Our data highlight the beneficial effects of these 5 LAB strains isolated from food in anti-infection and anti-inflammation.
Collapse
Affiliation(s)
- Rong Lu
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mei Shang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yong-Guo Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yang Jiao
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yinglin Xia
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Shari Garrett
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Danika Bakke
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Christine Bäuerl
- Lactic Acid Bacteria Laboratory, Department of Biotechnology, Instituto de Agroquimicay Tecnologia de Alimentos, Spanish National Research Council (C.S.I.C.), Valencia, Spain
| | - Gaspar Perez Martinez
- Lactic Acid Bacteria Laboratory, Department of Biotechnology, Instituto de Agroquimicay Tecnologia de Alimentos, Spanish National Research Council (C.S.I.C.), Valencia, Spain
| | - Cheol-Hyun Kim
- Animal Resource Science, Dankook University, Chungnam, Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Jun Sun
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,UIC Cancer Center, Chicago, Illinois, USA,Address correspondence to: Jun Sun, PhD, AGAF, FAPS, Professor, Division of Gastroenterology and Hepatology Department of Medicine, University of Illinois at Chicago 840 S. Wood Street, Room 704 CSB, MC716 Chicago, IL, 60612, USA. E-mail:
| |
Collapse
|
10
|
Cheng FS, Pan D, Chang B, Jiang M, Sang LX. Probiotic mixture VSL#3: An overview of basic and clinical studies in chronic diseases. World J Clin Cases 2020; 8:1361-1384. [PMID: 32368530 PMCID: PMC7190945 DOI: 10.12998/wjcc.v8.i8.1361] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 02/05/2023] Open
Abstract
Probiotics are known as “live microorganisms” and have been proven to have a health effect on hosts at the proper dose. Recently, a kind of probiotic mixture including eight live bacterial strains, VSL#3, has attracted considerable attention for its combined effect. VSL#3 is the only probiotic considered as a kind of medical food; it mainly participates in the regulation of the intestinal barrier function, including improving tight junction protein function, balancing intestinal microbial composition, regulating immune-related cytokine expression and so on. The objective of this review is to discuss the treatment action and mechanism for the administration of VSL#3 in chronic diseases of animals and humans (including children). We found that VSL#3 has a therapeutic or preventive effect in various systemic diseases per a large number of studies, including digestive systemic diseases (gastrointestinal diseases and hepatic diseases), obesity and diabetes, allergic diseases, nervous systemic diseases, atherosclerosis, bone diseases, and female reproductive systemic diseases.
Collapse
Affiliation(s)
- Fang-Shu Cheng
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
- Class 85 of 101k, China Medical University, Shenyang 110004, Liaoning Province, China
| | - Dan Pan
- Department of Geriatrics, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Chang
- Department of Gastroenterology, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Min Jiang
- Department of Gastroenterology, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Geriatrics, the First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
11
|
Jing B, Wang ZA, Zhang C, Deng Q, Wei J, Luo Y, Zhang X, Li J, Du Y. Establishment and Application of Peristaltic Human Gut-Vessel Microsystem for Studying Host-Microbial Interaction. Front Bioeng Biotechnol 2020; 8:272. [PMID: 32296697 PMCID: PMC7137556 DOI: 10.3389/fbioe.2020.00272] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
Intestinal floras influence a lot of biological functions of the organism. Although animal model are strong tools for researches on the relationship between host and microbe, a physiologically relevant in vitro human gut model was still required. Here, a novel human gut-vessel microfluidic system was established to study the host–microbial interaction. Peristaltic motion of the cells on the chip was driven by a pneumatic pump. When intestinal epithelial cells (Caco2) were co-cultured with vascular endothelial cells (HUVECs) on the peristaltic microfluidic chip, Caco2 showed normal barrier and absorption functions after 5 days cultivation, which generally took 21 days in static Transwell models. Intestinal microvilli and glycocalyx layer were seen after 4 days cultivation, and Lactobacillus casei was successfully co-cultured for a week in the intestinal cavity. A model for intestinal damage and inflammatory responses caused by E. coli was set up on this chip, which were successfully suppressed by Lactobacillus casei or antibiotic. In summary, this human gut-vessel microfluidic system showed a good potential for investigating the host–microbial interaction and the effect and mechanism of microbiome on intestinal diseases in vitro.
Collapse
Affiliation(s)
- Bolin Jing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China.,Department of Chemistry, University of Chinese Academy of Sciences, Beijing, China
| | - Zhuo A Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Chen Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Quanfeng Deng
- Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Jinhua Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yong Luo
- Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Xiuli Zhang
- College of Pharmaceutical Sciences, Soochow University, Soochow, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yuguang Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 1243] [Impact Index Per Article: 248.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Day RLJ, Harper AJ, Woods RM, Davies OG, Heaney LM. Probiotics: current landscape and future horizons. Future Sci OA 2019; 5:FSO391. [PMID: 31114711 PMCID: PMC6511921 DOI: 10.4155/fsoa-2019-0004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
In recent years there has been a rapid rise in interest for the application of probiotic supplements to act as mediators in health and disease. This appeal is predominantly due to ever-increasing evidence of the interaction of the microbiota and pathophysiological processes of disease within the human host. This narrative review considers the current landscape of the probiotic industry and its research, and discusses current pitfalls in the lack of translation from laboratory science to clinical application. Future considerations into how industry and academia must adapt probiotic research to maximize success are suggested, including more targeted application of probiotic strains dependent on individual capabilities as well as application of multiple advanced analytical technologies to further understand and accelerate microbiome science.
Collapse
Affiliation(s)
| | | | - Rachel M Woods
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Owen G Davies
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - Liam M Heaney
- School of Sport, Exercise & Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
14
|
Guo JR, Dong XF, Liu S, Tong JM. High-throughput sequencing reveals the effect of Bacillus subtilis CGMCC 1.921 on the cecal microbiota and gene expression in ileum mucosa of laying hens. Poult Sci 2018; 97:2543-2556. [PMID: 29897524 DOI: 10.3382/ps/pey112] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/09/2018] [Indexed: 12/16/2022] Open
Abstract
This study evaluated the effects of Bacillus subtilis CGMCC 1.921 supplementation on the production performance, cecal microbiota and mucosal transcriptome of laying hens by 16s rRNA gene sequencing and RNA-seq. A total of 144 27-week-old Hy-Line Brown laying hens were allocated into two treatments, namely, a basal diet without additions (T0) and the basal diet supplemented with 1.0 × 108 cfu/g (T1) B. subtilis CGMCC 1.921, with six replicates of 12 birds in each for 24 weeks. The results showed that T1 significantly decreased feed:egg ratio compared with T0 (P < 0.05). Dietary supplementation with B. subtilis CGMCC 1.921 increased the Shannon index (P < 0.05) which indicated enhanced diversity of cecal microflora. An increasing trend in Observed species index (P = 0.072) was observed in hens fed with diets supplemented with B. subtilis CGMCC 1.921 that showed a higher species richness. And T1 modulated cecal microbiota by increasing the relative proportion of Alistipes, Subdoligranulum, Ruminococcaceae UCG-014, Anaerotruncus, Ruminiclostridium 5, Ruminococcaceae UCG-010, Erysipelatoclostridium, Ruminococcaceae UCG-009, Family XIII AD3011 group, Bacillus, Faecalicoccus, Firmicutes bacterium CAG822, Oxalobacter, and Dielma at genus level (P < 0.05). In addition, there was a tendency of increase in the relative abundance of Lactobacillus (P = 0.055), Anaerobiospirillum (P = 0.059) and Family XIII UCG-001 (P = 0.054), Peptococcus (P = 0.078), and Ruminococcaceae UCG-004 (P = 0.078). Moreover, heatmap analysis indicated that the abundance of Campylobacter and Clostridium sensu stricto 1 was lower than T0. A total of 942 genes were identified by differential expression analysis, among which 400 genes were upregulated and 542 genes were downregulated. Bioinformatics analysis suggested that the upregulated genes were involved in Peroxisome Proliferator Activated Receptor (PPAR) signaling pathway, starch and sucrose metabolism, glycine/serine/threonine metabolism, and galactose metabolism, which may promote nutrient absorption. This study provided novel insights into the probiotic mechanisms of B. subtilis on laying hens.
Collapse
Affiliation(s)
- J R Guo
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - X F Dong
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - S Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - J M Tong
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| |
Collapse
|
15
|
Tomusiak-Plebanek A, Heczko P, Skowron B, Baranowska A, Okoń K, Thor PJ, Strus M. Lactobacilli with superoxide dismutase-like or catalase activity are more effective in alleviating inflammation in an inflammatory bowel disease mouse model. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3221-3233. [PMID: 30319243 PMCID: PMC6167993 DOI: 10.2147/dddt.s164559] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose Some lactobacilli, which possess superoxide dismutase-like activity and catalase activity naturally, have strong antioxidative properties. The aim of this study was to identify such strains and check which of them play a crucial role in alleviating intestinal inflammation. Methods We selected two Lactobacillus strains for use in animal studies: L. plantarum 30B (which has the highest catalase activity) and L. acidophilus 900 (which has the highest dismutase-like activity). Forty mice (C57B1/6J) were divided into four experimental groups with ten mice in each group. Group I (control group) was not supplemented with Lactobacillus, group II (catalase group) was orally supplemented with L. plantarum 30B, group III (dismutase-like group) was supplemented with L. acidophilus 900, and group IV (mixed group) was supplemented with both Lactobacillus strains. For 23 days, the temperature and body mass of each mouse were recorded and fecal samples for microbiological examination were collected. On day 23, the animals were sacrificed, and their intestines were removed for microbiological and histopathological studies. Results Compared to the control group, the highest drop in the body temperature was observed in groups II (P<0.05) and IV (P<0.05). Similarly, groups II (P<0.05) and IV (P<0.05) had the highest drop in body mass. Moreover, histopathological evaluation of colon fragments showed intracryptic abscesses in these groups. Group III mice showed most limited degree of inflammation. Conclusion Lactobacillus strains with dismutase-like activity are more effective in alleviating intestinal inflammation than strains producing catalase, suggesting that superoxide anion radical decomposition is crucial in this process.
Collapse
Affiliation(s)
| | - Piotr Heczko
- Department of Microbiology, Jagiellonian University Medical College, Poland,
| | - Beata Skowron
- Department of Pathophysiology, Jagiellonian University Medical College, Poland
| | | | - Krzysztof Okoń
- Department of Pathomorphology, Jagiellonian University Medical College, Poland
| | - Piotr J Thor
- Department of Pathophysiology, Jagiellonian University Medical College, Poland
| | - Magdalena Strus
- Department of Microbiology, Jagiellonian University Medical College, Poland,
| |
Collapse
|
16
|
A Synbiotic with Tumor Necrosis Factor- α Inhibitory Activity Ameliorates Experimental Jejunoileal Mucosal Injury. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9184093. [PMID: 29862296 PMCID: PMC5971273 DOI: 10.1155/2018/9184093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 03/31/2018] [Indexed: 02/06/2023]
Abstract
Despite the recent development of biological modifiers for inflammatory bowel diseases (IBD), there continues to be considerable interest in fermented medicines because of its negligible adverse effects. We previously showed that the synbiotic Gut Working Tablet (GWT) alleviates experimental colitis. Here we show that GWT is capable of ameliorating jejunoileal mucosal injury, which is frequently seen with IBD. We created experimental jejunoileal mucositis in rats by injection of methotrexate (MTX) which increases intestinal permeability, a hallmark finding of IBD. Administering GWT to MTX-injected rats restored intestinal integrity by reversing villi shortening, crypt loss, and goblet cell depletion in the mucosa. Also GWT reduced activities of myeloperoxidase and lipid peroxidase and increased superoxide dismutase activity, which is critical for maintaining intestinal function. We further found that GWT suppressed mRNA expression of tumor necrosis factor-α (TNF-α) and interleukin-12 (IL-12) in macrophage and reduced TNF-α mRNA expression in specimens with experimental colitis, which is in contrast to VSL#3 that enhanced TNF-α production. Together, the current and previous animal studies clearly demonstrate the protective role of GWT in chemically induced enterocolitis. Crohn's disease, a well-known IBD, can affect any portion of the intestine, and these results suggest that GWT may be useful as a novel therapeutic or maintenance therapy for IBD.
Collapse
|
17
|
Martín R, Chain F, Miquel S, Motta JP, Vergnolle N, Sokol H, Langella P. Using murine colitis models to analyze probiotics-host interactions. FEMS Microbiol Rev 2018; 41:S49-S70. [PMID: 28830096 DOI: 10.1093/femsre/fux035] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023] Open
Abstract
Probiotics are defined as 'live microorganisms which when administered in adequate amounts confer a health benefit on the host'. So, to consider a microorganism as a probiotic, a demonstrable beneficial effect on the health host should be shown as well as an adequate defined safety status and the capacity to survive transit through the gastrointestinal tract and to storage conditions. In this review, we present an overview of the murine colitis models currently employed to test the beneficial effect of the probiotic strains as well as an overview of the probiotics already tested. Our aim is to highlight both the importance of the adequate selection of the animal model to test the potential probiotic strains and of the value of the knowledge generated by these in vivo tests.
Collapse
Affiliation(s)
- Rebeca Martín
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Florian Chain
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Sylvie Miquel
- Laboratoire Microorganismes: Génome et Environnement (LMGE), UMR CNRS 6023, Université Clermont-Auvergne, 63000 Clermont-Ferrand, France
| | - Jean-Paul Motta
- Department of Biological Science, Inflammation Research Network, University of Calgary, AB T3E 4N1, Canada.,IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, F-31300 Toulouse, France
| | - Harry Sokol
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.,Sorbonne University - Université Pierre et Marie Curie (UPMC), 75252 Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) Equipe de Recherche Labélisée (ERL) 1157, Avenir Team Gut Microbiota and Immunity, 75012 Paris, France.,Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique - Hopitaux de Paris, UPMC, 75012 Paris, France
| | - Philippe Langella
- INRA, Commensals and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
18
|
Han X, Zhang J, Tan Y, Zhou G. Probiotics: A non-conventional therapy for oral lichen planus. Arch Oral Biol 2017; 81:90-96. [DOI: 10.1016/j.archoralbio.2017.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/10/2017] [Accepted: 04/21/2017] [Indexed: 12/14/2022]
|
19
|
Isidro RA, Lopez A, Cruz ML, Gonzalez Torres MI, Chompre G, Isidro AA, Appleyard CB. The Probiotic VSL#3 Modulates Colonic Macrophages, Inflammation, and Microflora in Acute Trinitrobenzene Sulfonic Acid Colitis. J Histochem Cytochem 2017; 65:445-461. [PMID: 28692320 PMCID: PMC5533272 DOI: 10.1369/0022155417718542] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/07/2017] [Indexed: 02/08/2023] Open
Abstract
The probiotic mixture VSL#3 attenuates colitis in patients with Inflammatory Bowel Disease (IBD) and in animal models of this condition, but the mechanisms involved are incompletely understood. VSL#3 alters macrophage morphology and secretory profile in vitro in a polarization-dependent manner. We examined the effect of VSL#3 on macrophages in acute trinitrobenzene sulfonic acid-induced colitis. Rats were randomized to normal, colitis, or colitis+VSL#3 groups. After sacrifice, the colons were evaluated for macroscopic and microscopic damage. Serum cytokine levels were measured, and microbiome analysis undertaken. Total and M1 colonic macrophages, and total and proliferating hepatic macrophages were assessed by double immunofluorescence staining. Colitis+VSL#3 rats had lower macroscopic damage, with less microscopic damage in the proximal colon, compared with colitis alone. Colitis significantly increased colonic macrophage infiltration, which was significantly reduced by VSL#3 treatment. VSL#3 did not decrease the colitis-induced surge of colonic M1 macrophages or hepatic macrophages. VSL#3 reduced colitis-induced serum cytokine levels, and induced restoration of colonic transcript levels for pro-inflammatory, anti-inflammatory, and barrier proteins to, or past, normal levels. Fecal bacteria distribution changed between groups. In summary, the probiotic VSL#3 reduces colitis severity, colonic macrophage infiltration, and serum cytokine levels, but does not dampen the pro-inflammatory phenotype of M1 macrophages.
Collapse
Affiliation(s)
- Raymond A. Isidro
- Raymond A. Isidro, Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, 395 Zona Industrial Reparada 2, Ponce, 00716, Puerto Rico. E-mail:
| | - Abdon Lopez
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| | - Myrella L. Cruz
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| | - Mayra I. Gonzalez Torres
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| | - Gladys Chompre
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| | - Angel A. Isidro
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| | - Caroline B. Appleyard
- Department of Basic Sciences, Medical School and Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico (RAI, MLC, AAI, CBA)
- Department of Biology, University of Puerto Rico at Ponce, Ponce, Puerto Rico (AL, MIGT)
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico (GC)
| |
Collapse
|
20
|
White R, Atherly T, Guard B, Rossi G, Wang C, Mosher C, Webb C, Hill S, Ackermann M, Sciabarra P, Allenspach K, Suchodolski J, Jergens AE. Randomized, controlled trial evaluating the effect of multi-strain probiotic on the mucosal microbiota in canine idiopathic inflammatory bowel disease. Gut Microbes 2017; 8:451-466. [PMID: 28678609 PMCID: PMC5628651 DOI: 10.1080/19490976.2017.1334754] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The intestinal microbiota is increasingly linked to the pathogenesis of idiopathic inflammatory bowel disease (IBD) in dogs. While studies have reported alterations in fecal (luminal) microbial populations, only limited information is available about the mucosal microbiota of IBD dogs at diagnosis and following medical therapy. Our aim was to characterize the mucosal microbiota and determine the clinical, microbiological, and mucosal homeostatic effects of probiotic treatment in dogs with IBD. Thirty four IBD dogs were randomized to receive standard therapy (ST = diet + prednisone) with or without probiotic. Tissue sections from endoscopic biopsies were evaluated by fluorescence in situ hybridization (FISH) on a quantifiable basis. Disease activity and changes in mucosal microbiota and tight junction protein (TJP) expression were assessed before and after 8 weeks of IBD therapy. ST and ST/probiotic therapy modulated the number of mucosal bacteria of IBD dogs in a similar fashion. Both treatments increased the numbers of total bacteria and individual species residing within adherent mucus, with ST therapy increasing Bifidobacterium spp. and ST/probiotic therapy increasing Lactobacillus spp (P < 0.05 for both), respectively. Both treatments were associated with rapid clinical remission but not improvement in histopathologic inflammation. Probiotic therapy was associated with upregulated (P < 0.05) expression of TJPs E-cadherin, occludin, and zonulin versus ST. The probiotic effect on mucosal bacteria is similar to that of IBD dogs receiving ST. IBD dogs fed probiotic had increased TJP expression suggesting that probiotic may have beneficial effects on mucosal homeostasis.
Collapse
Affiliation(s)
- Robin White
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | | | - Blake Guard
- Department of Small Animal Clinical Sciences, Gastrointestinal Laboratory, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Curtis Mosher
- Department of Genetics, Development & Cell Biology, College of Liberal Arts and Sciences, Iowa State University, Ames, Iowa, USA
| | - Craig Webb
- Colorado State University Veterinary Teaching Hospital, Fort Collins, CO, USA
| | - Steve Hill
- Veterinary Specialty Hospital – San Diego, CA, USA
| | - Mark Ackermann
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Peter Sciabarra
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Jan Suchodolski
- Department of Small Animal Clinical Sciences, Gastrointestinal Laboratory, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA,CONTACT Albert E. Jergens Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa 50011, USA
| |
Collapse
|
21
|
Shang M, Sun J. Vitamin D/VDR, Probiotics, and Gastrointestinal Diseases. Curr Med Chem 2017; 24:876-887. [PMID: 27915988 DOI: 10.2174/0929867323666161202150008] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/17/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022]
Abstract
Vitamin D is an important factor in regulating inflammation, immune responses, and carcinoma inhibition via action of its receptor, vitamin D receptor (VDR). Recent studies have demonstrated the role of vitamin D/VDR in regulating host-bacterial interactions. Probiotics are beneficial bacteria with the power of supporting or favoring life on the host. In the current review, we will discuss the recent progress on the roles of vitamin D/VDR in gut microbiome and inflammation. We will summarize evidence of probiotics in modulating vitamin D/VDR and balancing gut microbiota in health and gastrointestinal diseases. Moreover, we will review the clinical application of probiotics in prevention and therapy of IBD or colon cancer. Despite of the gains, there remain several barriers to advocate broad use of probiotics in clinical therapy. We will also discuss the limits and future direction in scientific understanding of probiotics, vitamin D/VDR, and host responses.
Collapse
Affiliation(s)
- Mei Shang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou. China
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, Chicago, IL, 60612. United States
| |
Collapse
|
22
|
Möhle L, Mattei D, Heimesaat MM, Bereswill S, Fischer A, Alutis M, French T, Hambardzumyan D, Matzinger P, Dunay IR, Wolf SA. Ly6C(hi) Monocytes Provide a Link between Antibiotic-Induced Changes in Gut Microbiota and Adult Hippocampal Neurogenesis. Cell Rep 2016; 15:1945-56. [PMID: 27210745 DOI: 10.1016/j.celrep.2016.04.074] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/15/2016] [Accepted: 04/20/2016] [Indexed: 12/18/2022] Open
Abstract
Antibiotics, though remarkably useful, can also cause certain adverse effects. We detected that treatment of adult mice with antibiotics decreases hippocampal neurogenesis and memory retention. Reconstitution with normal gut flora (SPF) did not completely reverse the deficits in neurogenesis unless the mice also had access to a running wheel or received probiotics. In parallel to an increase in neurogenesis and memory retention, both SPF-reconstituted mice that ran and mice supplemented with probiotics exhibited higher numbers of Ly6C(hi) monocytes in the brain than antibiotic-treated mice. Elimination of Ly6C(hi) monocytes by antibody depletion or the use of knockout mice resulted in decreased neurogenesis, whereas adoptive transfer of Ly6C(hi) monocytes rescued neurogenesis after antibiotic treatment. We propose that the rescue of neurogenesis and behavior deficits in antibiotic-treated mice by exercise and probiotics is partially mediated by Ly6C(hi) monocytes.
Collapse
Affiliation(s)
- Luisa Möhle
- Institute of Medical Microbiology, University of Magdeburg, 39106 Magdeburg, Germany
| | - Daniele Mattei
- Department of Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Markus M Heimesaat
- Charité - University Medicine Berlin, Department of Microbiology and Hygiene, 14195 Berlin, Germany
| | - Stefan Bereswill
- Charité - University Medicine Berlin, Department of Microbiology and Hygiene, 14195 Berlin, Germany
| | - André Fischer
- Charité - University Medicine Berlin, Department of Microbiology and Hygiene, 14195 Berlin, Germany
| | - Marie Alutis
- Charité - University Medicine Berlin, Department of Microbiology and Hygiene, 14195 Berlin, Germany
| | - Timothy French
- Institute of Medical Microbiology, University of Magdeburg, 39106 Magdeburg, Germany
| | - Dolores Hambardzumyan
- Department of Neurosciences at the Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA; Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Polly Matzinger
- Ghost Lab, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892-9760, USA
| | - Ildiko R Dunay
- Institute of Medical Microbiology, University of Magdeburg, 39106 Magdeburg, Germany
| | - Susanne A Wolf
- Department of Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| |
Collapse
|
23
|
Vasconcelos RM, Sanfilippo N, Paster BJ, Kerr AR, Li Y, Ramalho L, Queiroz EL, Smith B, Sonis ST, Corby PM. Host-Microbiome Cross-talk in Oral Mucositis. J Dent Res 2016; 95:725-33. [PMID: 27053118 DOI: 10.1177/0022034516641890] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oral mucositis (OM) is among the most common, painful, and debilitating toxicities of cancer regimen-related treatment, resulting in the formation of ulcers, which are susceptible to increased colonization of microorganisms. Novel discoveries in OM have focused on understanding the host-microbial interactions, because current pathways have shown that major virulence factors from microorganisms have the potential to contribute to the development of OM and may even prolong the existence of already established ulcerations, affecting tissue healing. Additional comprehensive and disciplined clinical investigation is needed to carefully characterize the relationship between the clinical trajectory of OM, the local levels of inflammatory changes (both clinical and molecular), and the ebb and flow of the oral microbiota. Answering such questions will increase our knowledge of the mechanisms engaged by the oral immune system in response to mucositis, facilitating their translation into novel therapeutic approaches. In doing so, directed clinical strategies can be developed that specifically target those times and tissues that are most susceptible to intervention.
Collapse
Affiliation(s)
- R M Vasconcelos
- School of Medicine, New York University, New York, NY, USA College of Dentistry, New York University, New York, NY, USA Faculdade de Odontologia, Universidade Federal da Bahia, Salvador, BA, Brazil
| | - N Sanfilippo
- School of Medicine, New York University, New York, NY, USA New York University Perlmutter Cancer Center, New York, NY, USA
| | - B J Paster
- The Forsyth Institute, Cambridge, MA, USA Department of Oral Medicine, Infection & Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - A R Kerr
- College of Dentistry, New York University, New York, NY, USA
| | - Y Li
- College of Dentistry, New York University, New York, NY, USA
| | - L Ramalho
- Faculdade de Odontologia, Universidade Federal da Bahia, Salvador, BA, Brazil
| | - E L Queiroz
- College of Dentistry, New York University, New York, NY, USA
| | - B Smith
- School of Medicine, New York University, New York, NY, USA New York University Perlmutter Cancer Center, New York, NY, USA
| | - S T Sonis
- Biomodels, LLC, Watertown, MA, USA Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - P M Corby
- School of Medicine, New York University, New York, NY, USA College of Dentistry, New York University, New York, NY, USA
| |
Collapse
|
24
|
Ranji P, Akbarzadeh A, Rahmati-Yamchi M. Associations of Probiotics with Vitamin D and Leptin Receptors and their Effects on Colon Cancer. Asian Pac J Cancer Prev 2016; 16:3621-7. [PMID: 25987012 DOI: 10.7314/apjcp.2015.16.9.3621] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Colorectal cancer (CRC) is one of most common causes of cancer-related death worldwide. Recent studies have suggested that microbial and environmental factors including diet and lifestyle can impact on colon cancer development. Vitamin D deficiency and dysfunction of vitamin D receptor (VDR) also correlate with colon cancer. Moreover, leptin, a 16-kDa polypeptide, participates in the regulation of food intake and is associated with other environmental factors affecting colon cancer through the leptin receptor. Altered levels of serum leptin and patterns of expression of its receptor (LPR) may be observed in human colon tumours. Furthermore, the collected data from in vitro and in vivo studies have indicated that consuming probiotic non-pathogenic lactic acid bacteria have beneficial effects on colon cancer. Probiotics, inflammation and vitamin D/VDR have been correlated with leptin and its receptor and are also with colon cancer. Thus, in this paper, we review recent progress on the roles of probiotic, vitamin D/VDR and leptin/LPR in inflammation and colon cancer.
Collapse
Affiliation(s)
- Peyman Ranji
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, International Branch of Tabriz University of Medical sciences (Aras), Tabriz, Iran E-mail :
| | | | | |
Collapse
|
25
|
Ferguson LR. Fish oils in parenteral nutrition: Why could these be important for gastrointestinal oncology? World J Gastrointest Oncol 2015; 7:128-131. [PMID: 26380055 PMCID: PMC4569589 DOI: 10.4251/wjgo.v7.i9.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/07/2015] [Indexed: 02/05/2023] Open
Abstract
By the time a gastroenterology patient is moved to parenteral nutrition, he or she is usually in poor health. All parenteral nutrition formulae contain essential nutrients, avoiding components that could cause an adverse reaction. The lipid component is often provided by a soy extract, containing all the fatty acids considered to be essential in the diet. Several trials have considered parenteral nutrition formulas with added fish oils, high in the long chain omega-3 polyunsaturated fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Given the range of biological functions associated with such compounds, especially in reducing inflammatory symptoms, this move would appear rational. However, while data from such trials are often positive, there has been variability among results. Some of this variability could be caused by environmental contaminants in the fish, and/or oxidation of the lipids because of poor storage. The situation is complicated by a recent report that fish oils may counter the effects of platinum chemotherapy. However, this effect associated with a minor component, hexadeca-4,7,10,13-tetraenoic acid. It is suggested that pure DHA and EPA would be beneficial additions to parenteral nutrition, reducing the probability of carcinogenesis and enhancing rational disease management. However, the jury is still out on fish oils more generally.
Collapse
|
26
|
Wu S, Yoon S, Zhang YG, Lu R, Xia Y, Wan J, Petrof EO, Claud EC, Chen D, Sun J. Vitamin D receptor pathway is required for probiotic protection in colitis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G341-9. [PMID: 26159695 PMCID: PMC4556945 DOI: 10.1152/ajpgi.00105.2015] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/26/2015] [Indexed: 02/08/2023]
Abstract
Low expression of vitamin D receptor (VDR) and dysfunction of vitamin D/VDR signaling are reported in patients with inflammatory bowel disease (IBD); therefore, restoration of VDR function to control inflammation in IBD is desirable. Probiotics have been used in the treatment of IBD. However, the role of probiotics in the modulation of VDR signaling to effectively reduce inflammation is unknown. We identified a novel role of probiotics in activating VDR activity, thus inhibiting inflammation, using cell models and VDR knockout mice. We found that the probiotics Lactobacillus rhamnosus strain GG (LGG) and Lactobacillus plantarum (LP) increased VDR protein expression in both mouse and human intestinal epithelial cells. Using the VDR luciferase reporter vector, we detected increased transcriptional activity of VDR after probiotic treatment. Probiotics increased the expression of the VDR target genes, such as antimicrobial peptide cathelicidin, at the transcriptional level. Furthermore, the role of probiotics in regulating VDR signaling was tested in vivo using a Salmonella-colitis model in VDR knockout mice. Probiotic treatment conferred physiological and histologic protection from Salmonella-induced colitis in VDR(+/+) mice, whereas probiotics had no effects in the VDR(-/-) mice. Probiotic treatment also enhanced numbers of Paneth cells, which secrete AMPs for host defense. These data indicate that the VDR pathway is required for probiotic protection in colitis. Understanding how probiotics enhance VDR signaling and inhibit inflammation will allow probiotics to be used effectively, resulting in innovative approaches to the prevention and treatment of chronic inflammation.
Collapse
Affiliation(s)
- Shaoping Wu
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Sonia Yoon
- 2Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medical College, Cornell University, New York, New York;
| | - Yong-Guo Zhang
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Rong Lu
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Yinglin Xia
- 3Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York;
| | - Jiandi Wan
- 4Microsystems Engineering, Rochester Institute of Technology, Rochester, New York;
| | - Elaine O. Petrof
- 5Department of Medicine, Gastrointestinal Diseases Research Unit and Division of Infectious Diseases, Queen's University, Kingston, Ontario, Canada; and
| | - Erika C. Claud
- 6Departments of Pediatrics and Medicine, The University of Chicago Medical Center, Chicago, Illinois
| | - Di Chen
- 1Department of Biochemistry, Rush University, Chicago, Illinois;
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, Illinois;
| |
Collapse
|
27
|
Abstract
BACKGROUND Current understanding of the onset of inflammatory bowel diseases relies heavily on data derived from animal models of colitis. However, the omission of information concerning the method used makes the interpretation of studies difficult or impossible. We assessed the current quality of methods reporting in 4 animal models of colitis that are used to inform clinical research into inflammatory bowel disease: dextran sulfate sodium, interleukin-10, CD45RB T cell transfer, and 2,4,6-trinitrobenzene sulfonic acid (TNBS). METHODS We performed a systematic review based on PRISMA guidelines, using a PubMed search (2000-2014) to obtain publications that used a microarray to describe gene expression in colitic tissue. Methods reporting quality was scored against a checklist of essential and desirable criteria. RESULTS Fifty-eight articles were identified and included in this review (29 dextran sulfate sodium, 15 interleukin-10, 5 T cell transfer, and 16 TNBS; some articles use more than 1 colitis model). A mean of 81.7% (SD = ±7.038) of criteria were reported across all models. Only 1 of the 58 articles reported all essential criteria on our checklist. Animal age, gender, housing conditions, and mortality/morbidity were all poorly reported. CONCLUSIONS Failure to include all essential criteria is a cause for concern; this failure can have large impact on the quality and replicability of published colitis experiments. We recommend adoption of our checklist as a requirement for publication to improve the quality, comparability, and standardization of colitis studies and will make interpretation and translation of data to human disease more reliable.
Collapse
|
28
|
Glenn AJ, Fielding KA, Chen J, Comelli EM, Ward WE. Long-term vitamin D3 supplementation does not prevent colonic inflammation or modulate bone health in IL-10 knockout mice at young adulthood. Nutrients 2014; 6:3847-62. [PMID: 25247786 PMCID: PMC4179191 DOI: 10.3390/nu6093847] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/04/2014] [Accepted: 09/09/2014] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic disease that can impair bone metabolism. Low vitamin D status has been implicated in its progress. This study used interleukin (IL)-10 knockout (KO) mice, that develop an intestinal inflammation when housed in a non-sterile environment, to determine if supplementation with vitamin D3 throughout life could mitigate inflammation and attenuate the lower bone mineral content (BMC) and density (BMD), and bone strength. Female IL-10 KO mice were randomized 25 or 5000 IU vitamin D3/kg diet throughout pregnancy and lactation. At weaning, offspring received the same or opposite diet as their mother until age three months. Body weight growth was similar among groups within a sex. At three months of age, there were no differences in inflammation and gene expression in the colon of offspring. Male offspring exposed to continuous 25 IU vitamin D3/kg diet had lower (p < 0.001) colonic VDR expression and those exposed only to low vitamin D3 until weaning had higher serum IL-6. There were no differences in femur or vertebral BMC, BMD or bone strength. In summary, long-term exposure to vitamin D3 did not attenuate intestinal inflammation or preserve bone mineral or bone strength. Thus, supplementation with vitamin D3 does not exert anti-inflammatory effects in this mouse model that mimics human inflammatory bowel disease.
Collapse
Affiliation(s)
- Andrea J Glenn
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada.
| | | | - Jianmin Chen
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada.
| | - Elena M Comelli
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada.
| | - Wendy E Ward
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada.
| |
Collapse
|
29
|
Distrutti E, O’Reilly JA, McDonald C, Cipriani S, Renga B, Lynch MA, Fiorucci S. Modulation of intestinal microbiota by the probiotic VSL#3 resets brain gene expression and ameliorates the age-related deficit in LTP. PLoS One 2014; 9:e106503. [PMID: 25202975 PMCID: PMC4159266 DOI: 10.1371/journal.pone.0106503] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/19/2014] [Indexed: 12/17/2022] Open
Abstract
The intestinal microbiota is increasingly recognized as a complex signaling network that impacts on many systems beyond the enteric system modulating, among others, cognitive functions including learning, memory and decision-making processes. This has led to the concept of a microbiota-driven gut–brain axis, reflecting a bidirectional interaction between the central nervous system and the intestine. A deficit in synaptic plasticity is one of the many changes that occurs with age. Specifically, the archetypal model of plasticity, long-term potentiation (LTP), is reduced in hippocampus of middle-aged and aged rats. Because the intestinal microbiota might change with age, we have investigated whether the age-related deficit in LTP might be attenuated by changing the composition of intestinal microbiota with VSL#3, a probiotic mixture comprising 8 Gram-positive bacterial strains. Here, we report that treatment of aged rats with VSL#3 induced a robust change in the composition of intestinal microbiota with an increase in the abundance of Actinobacteria and Bacterioidetes, which was reduced in control-treated aged rats. VSL#3 administration modulated the expression of a large group of genes in brain tissue as assessed by whole gene expression, with evidence of a change in genes that impact on inflammatory and neuronal plasticity processes. The age-related deficit in LTP was attenuated in VSL#3-treated aged rats and this was accompanied by a modest decrease in markers of microglial activation and an increase in expression of BDNF and synapsin. The data support the notion that intestinal microbiota can be manipulated to positively impact on neuronal function.
Collapse
Affiliation(s)
- Eleonora Distrutti
- S.C. di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
- * E-mail:
| | - Julie-Ann O’Reilly
- Trinity College Institute for Neuroscience, Department of Physiology, Trinity College, Dublin, Ireland
| | - Claire McDonald
- Trinity College Institute for Neuroscience, Department of Physiology, Trinity College, Dublin, Ireland
| | - Sabrina Cipriani
- Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Barbara Renga
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| | - Marina A. Lynch
- Trinity College Institute for Neuroscience, Department of Physiology, Trinity College, Dublin, Ireland
| | - Stefano Fiorucci
- Dipartimento di Scienze Chirurgiche e Biomediche, Università degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
30
|
Zarrati M, Salehi E, Nourijelyani K, Mofid V, Zadeh MJH, Najafi F, Ghaflati Z, Bidad K, Chamari M, Karimi M, Shidfar F. Effects of probiotic yogurt on fat distribution and gene expression of proinflammatory factors in peripheral blood mononuclear cells in overweight and obese people with or without weight-loss diet. J Am Coll Nutr 2014; 33:417-25. [PMID: 25079040 DOI: 10.1080/07315724.2013.874937] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate whether probiotics had an effect on proinflammatory markers and cytokines in overweight and obese individuals and whether they could have synergistic effects with weight-loss diets. METHODS A total of 75 healthy overweight and obese individuals completed this randomized doubled-blind controlled clinical trial. Participants were randomly assigned to groups consuming regular yogurt with a low-calorie diet (LCD, RLCD; n = 25) or receiving probiotic yogurt with LCD (PLCD; n = 25) or consuming probiotic yogurt without LCD (PWLCD; n = 25) for 8 weeks. The pribiotic regimen contained 200 g/day yogurt, enriched by Lactobacillus acidophilus La5, Bifidobacterium BB12, and Lactobacillus casei DN001 10(8) colony-forming units/g. Body fat percentage, high-sensitive C-reactive protein (hs-CRP), tumor necrosis factor-alpha (TNF-α), leptin, and mRNA levels of inflammation-related genes (TNF-α and RAR-related orphan receptor gamma [ROR-γt]) in peripheral blood mononuclear cells (PBMCs) were measured. RESULTS A reduction in body mass index (BMI), fat percentage, and leptin level was observed that was more obvious in groups who received the weight-loss diet with probiotic yogurt. Reduction in the gene expression of ROR-γt was significant in the PLCD group (p < 0.001). The expression of TNF-α did not change among all groups after intervention. The mean concentration of leptin was significantly decreased in all groups after the dietary intervention, but the mean changes in leptin level in the PLCD group was more prominent compared to the other two groups (-2.38, p < 0.001 [PLCD] vs -1.75, p = 0.002 [RLCD] and -0.55 ng/mL, p = 0.12 [PWLCD]). The reduction in serum levels of hs-CRP was more evident in the PWLCD group compared to the PLCD and RLCD groups after the 8-week intervention (-3.4, p = 0.03 vs -1.76, p < 0.001 and -2.98 pg/mL, p < 0.001, respectively). CONCLUSION Our results suggested that the weight-loss diet and probiotic yogurt had synergistic effects on T-cells subset specific gene expression in PBMCs, fat percentage, and body weight among overweight and obese individuals.
Collapse
Affiliation(s)
- Mitra Zarrati
- a Department of Nutrition, School of Health , Tehran University of Medical Sciences , Tehran , IRAN
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Martini S, Nair V, Keller BJ, Eichinger F, Hawkins JJ, Randolph A, Böger CA, Gadegbeku CA, Fox CS, Cohen CD, Kretzler M. Integrative biology identifies shared transcriptional networks in CKD. J Am Soc Nephrol 2014; 25:2559-72. [PMID: 24925724 DOI: 10.1681/asn.2013080906] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A previous meta-analysis of genome-wide association data by the Cohorts for Heart and Aging Research in Genomic Epidemiology and CKDGen consortia identified 16 loci associated with eGFR. To define how each of these single-nucleotide polymorphisms (SNPs) could affect renal function, we integrated GFR-associated loci with regulatory pathways, producing a molecular map of CKD. In kidney biopsy specimens from 157 European subjects representing nine different CKDs, renal transcript levels for 18 genes in proximity to the SNPs significantly correlated with GFR. These 18 genes were mapped into their biologic context by testing coregulated transcripts for enriched pathways. A network of 97 pathways linked by shared genes was constructed and characterized. Of these pathways, 56 pathways were reported previously to be associated with CKD; 41 pathways without prior association with CKD were ranked on the basis of the number of candidate genes connected to the respective pathways. All pathways aggregated into a network of two main clusters comprising inflammation- and metabolism-related pathways, with the NRF2-mediated oxidative stress response pathway serving as the hub between the two clusters. In all, 78 pathways and 95% of the connections among those pathways were verified in an independent North American biopsy cohort. Disease-specific analyses showed that most pathways are shared between sets of three diseases, with closest interconnection between lupus nephritis, IgA nephritis, and diabetic nephropathy. Taken together, the network integrates candidate genes from genome-wide association studies into their functional context, revealing interactions and defining established and novel biologic mechanisms of renal impairment in renal diseases.
Collapse
Affiliation(s)
- Sebastian Martini
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Viji Nair
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Benjamin J Keller
- Department of Computer Science, Eastern Michigan University, Ypsilanti, Michigan
| | - Felix Eichinger
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jennifer J Hawkins
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Ann Randolph
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Carsten A Böger
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Crystal A Gadegbeku
- Department of Medicine, Section of Nephrology and Kidney Transplantation, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Caroline S Fox
- Division of Intramural Research and Laboratory for Population and Metabolic Health, National Heart, Lung, and Blood Institute, Framingham, Massachusetts; Department of Endocrinology, Brigham and Women's Hospital, Boston, Massachusetts; and
| | - Clemens D Cohen
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Matthias Kretzler
- Departments of Internal Medicine, Nephrology, and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan;
| | | | | | | |
Collapse
|
32
|
Noto Llana M, Sarnacki SH, Aya Castañeda MDR, Bernal MI, Giacomodonato MN, Cerquetti MC. Consumption of Lactobacillus casei fermented milk prevents Salmonella reactive arthritis by modulating IL-23/IL-17 expression. PLoS One 2013; 8:e82588. [PMID: 24340048 PMCID: PMC3858332 DOI: 10.1371/journal.pone.0082588] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/25/2013] [Indexed: 12/11/2022] Open
Abstract
Reactive arthritis is the development of sterile joint inflammation as a sequel to a remote infection, often in the gut. We have previously shown that a low dose of S. enteritidis inoculated to streptomycin-pretreated mice generates a self-limiting enterocolitis suitable for studying reactive arthritis. Here we show that consumption of Lactobacillus casei prior to infection abolishes intestinal and joint inflammation triggered by Salmonella. BALB/c mice were sacrificed after infection; intestinal and joint samples were analyzed for histological changes and expression of cytokines. TNF-α was measured by ELISA and the expression of IL-1β, IL-6, IL-10, IL-17, IL-23 and TGF-β was assessed by qPCR. L. casei consumption prevented Salmonella-induced synovitis, the increment of TNF-α in knees and the increase of IL-17 expression in popliteal and inguinal lymph nodes. At intestinal level consumption of L. casei drastically diminished S. enteritidis invasiveness and shortened splenic persistence of the pathogen. Bacterial loads recovered at days 2 and 5 from Peyer's patches were 10-fold lower in mice fed with L. casei. In accordance, we found that the augment in gut permeability induced during enterocolitis was decreased in those animals. Consumption of L. casei prior to infection failed to increase anti- inflammatory molecules such as IL-10 and TGF-β in the intestine. On the other hand, consumption of L. casei abrogated the expression of TNF-α, IL-17, IL-23, IL-1β and IL-6 in cecum and mesenteric lymph nodes. These cytokines are needed for differentiation of immune cells involved in the development of reactive arthritis such as Th17 and γδ T cells. Trafficking of these inflammatory cells from the gut to the joints has been proposed as a mechanism of generation of reactive arthritis. Our results suggest that L. casei consumption prevents Salmonella-induced synovitis by altering the intestinal milieu necessary for differentiation of cells involved in the generation of joint inflammation.
Collapse
Affiliation(s)
- Mariángeles Noto Llana
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sebastián Hernán Sarnacki
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María del Rosario Aya Castañeda
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Isabel Bernal
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica Nancy Giacomodonato
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Cristina Cerquetti
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
33
|
Arthur JC, Gharaibeh RZ, Uronis JM, Perez-Chanona E, Sha W, Tomkovich S, Mühlbauer M, Fodor AA, Jobin C. VSL#3 probiotic modifies mucosal microbial composition but does not reduce colitis-associated colorectal cancer. Sci Rep 2013; 3:2868. [PMID: 24100376 PMCID: PMC3792409 DOI: 10.1038/srep02868] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 09/18/2013] [Indexed: 02/07/2023] Open
Abstract
Although probiotics have shown success in preventing the development of experimental colitis-associated colorectal cancer (CRC), beneficial effects of interventional treatment are relatively unknown. Here we show that interventional treatment with VSL#3 probiotic alters the luminal and mucosally-adherent microbiota, but does not protect against inflammation or tumorigenesis in the azoxymethane (AOM)/Il10⁻/⁻ mouse model of colitis-associated CRC. VSL#3 (10⁹ CFU/animal/day) significantly enhanced tumor penetrance, multiplicity, histologic dysplasia scores, and adenocarcinoma invasion relative to VSL#3-untreated mice. Illumina 16S sequencing demonstrated that VSL#3 significantly decreased (16-fold) the abundance of a bacterial taxon assigned to genus Clostridium in the mucosally-adherent microbiota. Mediation analysis by linear models suggested that this taxon was a contributing factor to increased tumorigenesis in VSL#3-fed mice. We conclude that VSL#3 interventional therapy can alter microbial community composition and enhance tumorigenesis in the AOM/Il10⁻/⁻ model.
Collapse
Affiliation(s)
| | - Raad Z. Gharaibeh
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, NC 28081, USA
| | | | | | - Wei Sha
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, NC 28081, USA
| | - Sarah Tomkovich
- Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Anthony A. Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Christian Jobin
- Department of Medicine, Chapel Hill, NC 27599, USA
- Pharmacology, Chapel Hill, NC 27599, USA
- Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of Florida at Gainesville, Gainesville, FL32611, USA
- Department of Infectious Diseases and Pathology, University of Florida at Gainesville, Gainesville, FL32611, USA
| |
Collapse
|
34
|
Gawkowski D, Chikindas M. Non-dairy probiotic beverages: the next step into human health. Benef Microbes 2013; 4:127-42. [DOI: 10.3920/bm2012.0030] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, confer a health benefit to the host. The two main genera of microorganisms indicated as sources of probiotic bacteria are Lactobacillus and Bifidobacterium. Historically used to produce fermented dairy products, certain strains of both genera are increasingly utilised to formulate other functional foods. As the consumers’ understanding of the role of probiotics in health grows, so does the popularity of food containing them. The result of this phenomenon is an increase in the number of probiotic foods available for public consumption, including a rapidly-emerging variety of probiotic-containing non-dairy beverages, which provide a convenient way to improve and maintain health. However, the composition of non-dairy probiotic beverages can pose specific challenges to the survival of the health conferring microorganisms. To overcome these challenges, strain selection and protection techniques play an integral part in formulating a stable product. This review discusses non-dairy probiotic beverages, characteristics of an optimal beverage, and commonly used probiotic strains, including spore-forming bacteria. It also examines the most recent developments in probiotic encapsulation technology with focus on nano-fibre formation as a means of protecting viable cells. Utilising bacteria's natural armour or creating barrier mechanisms via encapsulation technology will fuel development of stable non-dairy probiotic beverages.
Collapse
Affiliation(s)
- D. Gawkowski
- School of Environmental and Biological Sciences, Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - M.L. Chikindas
- School of Environmental and Biological Sciences, Department of Food Science, Rutgers, The State University of New Jersey, 65 Dudley Road, New Brunswick, NJ 08901, USA
| |
Collapse
|
35
|
Effect of Lactobacillus brevis KB290 on the cell-mediated cytotoxic activity of mouse splenocytes: a DNA microarray analysis. Br J Nutr 2013; 110:1617-29. [PMID: 23544404 DOI: 10.1017/s0007114513000767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lactic acid bacteria confer a variety of health benefits. Here, we investigate the mechanisms by which Lactobacillus brevis KB290 (KB290) enhances cell-mediated cytotoxic activity. Female BALB/c mice aged 9 weeks were fed a diet containing KB290 (3 × 10(9) colony-forming units/g) or starch for 1 d. The resulting cytotoxic activity of splenocytes against YAC-1 cells was measured using flow cytometry and analysed for gene expression using DNA microarray technology. KB290 enhanced the cell-mediated cytotoxic activity of splenocytes. DNA microarray analysis identified 327 up-regulated and 347 down-regulated genes that characterised the KB290 diet group. The up-regulated genes were significantly enriched in Gene Ontology terms related to immunity, and, especially, a positive regulation of T-cell-mediated cytotoxicity existed among these terms. Almost all the genes included in the term encoded major histocompatibility complex (MHC) class I molecules involved in the presentation of antigen to CD8(+) cytotoxic T cells. Marco and Signr1 specific to marginal zone macrophages (MZM), antigen-presenting cells, were also up-regulated. Flow cytometric analysis confirmed that the proportion of MZM was significantly increased by KB290 ingestion. Additionally, the over-represented Kyoto Encyclopedia of Genes and Genomes pathways among the up-regulated genes were those for natural killer (NK) cell-mediated cytotoxicity and antigen processing and presentation. The results for the selected genes associated with NK cells and CD8(+) cytotoxic T cells were confirmed by quantitative RT-PCR. These results suggest that enhanced cytotoxic activity could be caused by the activation of NK cells and/or of CD8(+) cytotoxic T cells stimulated via MHC class I presentation.
Collapse
|
36
|
Ortuño Sahagún D, Márquez-Aguirre AL, Quintero-Fabián S, López-Roa RI, Rojas-Mayorquín AE. Modulation of PPAR-γ by Nutraceutics as Complementary Treatment for Obesity-Related Disorders and Inflammatory Diseases. PPAR Res 2012; 2012:318613. [PMID: 23251142 PMCID: PMC3515933 DOI: 10.1155/2012/318613] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/03/2012] [Accepted: 10/23/2012] [Indexed: 12/16/2022] Open
Abstract
A direct correlation between adequate nutrition and health is a universally accepted truth. The Western lifestyle, with a high intake of simple sugars, saturated fat, and physical inactivity, promotes pathologic conditions. The main adverse consequences range from cardiovascular disease, type 2 diabetes, and metabolic syndrome to several cancers. Dietary components influence tissue homeostasis in multiple ways and many different functional foods have been associated with various health benefits when consumed. Natural products are an important and promising source for drug discovery. Many anti-inflammatory natural products activate peroxisome proliferator-activated receptors (PPAR); therefore, compounds that activate or modulate PPAR-gamma (PPAR-γ) may help to fight all of these pathological conditions. Consequently, the discovery and optimization of novel PPAR-γ agonists and modulators that would display reduced side effects is of great interest. In this paper, we present some of the main naturally derived products studied that exert an influence on metabolism through the activation or modulation of PPAR-γ, and we also present PPAR-γ-related diseases that can be complementarily treated with nutraceutics from functional foods.
Collapse
Affiliation(s)
- D. Ortuño Sahagún
- Laboratorio de Desarrollo y Regeneración Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, camino Ing. R. Padilla Sánchez 2100, Las Agujas, 44600 Zapopan JAL, Mexico
| | - A. L. Márquez-Aguirre
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., 44270 Guadalajara, JAL, Mexico
| | - S. Quintero-Fabián
- Laboratorio de Desarrollo y Regeneración Neural, Instituto de Neurobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, camino Ing. R. Padilla Sánchez 2100, Las Agujas, 44600 Zapopan JAL, Mexico
- Departamento de Farmacobiología, CUCEI, Universidad de Guadalajara, Boulevard Marcelino García Barragán, 44430 Tlaquepaque, JAL, Mexico
| | - R. I. López-Roa
- Departamento de Farmacobiología, CUCEI, Universidad de Guadalajara, Boulevard Marcelino García Barragán, 44430 Tlaquepaque, JAL, Mexico
| | - A. E. Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, 45100, JAL, Mexico
- Departamento de Investigación Básica, Instituto Nacional de Geriatría (INGER), Periférico Sur No. 2767, Col, San Jerónimo Lídice, Delegación Magdalena Contreras 10200, México DF, Mexico
| |
Collapse
|
37
|
Nanau RM, Neuman MG. Nutritional and probiotic supplementation in colitis models. Dig Dis Sci 2012; 57:2786-810. [PMID: 22736018 DOI: 10.1007/s10620-012-2284-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/08/2012] [Indexed: 01/01/2023]
Abstract
In vitro and animals models have long been used to study human diseases and identify novel therapeutic approaches that can be applied to combat these conditions. Ulcerative colitis and Crohn's disease are the two main entities of inflammatory bowel disease (IBD). There is an intricate relationship between IBD features in human patients, in vitro and animal colitis models, mechanisms and possible therapeutic approaches in these models, and strategies that can be extrapolated and applied in humans. Malnutrition, particularly protein-energy malnutrition and vitamin and micronutrient deficiencies, as well as dysregulation of the intestinal microbiota, are common features of IBD. Based on these observations, dietary supplementation with essential nutrients known to be in short supply in the diet in IBD patients and with other molecules believed to provide beneficial anti-inflammatory effects, as well as with probiotic organisms that stimulate immune functions and resistance to infection has been tested in colitis models. Here we review current knowledge on nutritional and probiotic supplementation in in vitro and animal colitis models. While some of these strategies require further fine-tuning before they can be applied in human IBD patients, their intended purpose is to prevent, delay or treat disease symptoms in a non-pharmaceutical manner.
Collapse
Affiliation(s)
- Radu M Nanau
- Department of Pharmacology and Toxicology, Institute of Drug Research, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
38
|
Abstract
The healthy human gut supports a complex and diverse microbiota, dominated by bacterial phylotypes belonging to Bacteroidetes and Firmicutes. In the inflamed gut, overall diversity decreases, coincident with a greater representation of Proteobacteria. There is growing evidence supporting an important role for human gut bacteria in mucosal immunity; interactions at the level of both intestinal and colonic epithelial cells, dendritic cells, and T and B immune cells have been documented. These interactions influence gut barrier and defense mechanisms that include antimicrobial peptide and secretory IgA synthesis. The functional effects of commensal bacteria on T helper cell differentiation have led to the emerging concept that microbiota composition determines T effector- and T regulatory-cell balance, immune responsiveness, and homeostasis. The importance of this biology in relation to immune homeostasis, inflammatory bowel disease, and the rising incidence of autoimmune diseases will be discussed. The detailed description of the human gut microbiota, integrated with evidence-based mechanisms of immune modulation, provides an exciting platform for the identification of next-generation probiotics and related pharmaceutical products.
Collapse
Affiliation(s)
- Denise Kelly
- Rowett Institute of Nutrition & Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK.
| | | |
Collapse
|
39
|
Mencarelli A, Cipriani S, Renga B, Bruno A, D'Amore C, Distrutti E, Fiorucci S. VSL#3 resets insulin signaling and protects against NASH and atherosclerosis in a model of genetic dyslipidemia and intestinal inflammation. PLoS One 2012; 7:e45425. [PMID: 23029000 PMCID: PMC3448636 DOI: 10.1371/journal.pone.0045425] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/16/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Signals generated by the inflammed intestine are thought to contribute to metabolic derangement. The intestinal microbiota contributes to instructing the immune system beyond the intestinal wall and its modulation is a potential target for treating systemic disorders. AIMS To investigate the pathogenetic role of low grade intestinal inflammation in the development of steatohepatitis and atherosclerosis in a model of genetic dyslipidemia and to test the therapeutic potential of a probiotics intervention in protecting against development of these disorders. RESULTS ApoE(-/-) mice were randomized to receive vehicle or VSL#3, a mixture of eight probiotics, at the dose of 20×10(9) colony-forming units/kg/day for three months alone or in combination with 0.2% of dextran sulfate sodium (DSS) in drinking water. Administering DSS to ApoE(-/-) mice failed to induce signs and symptoms of colitis but increased intestinal permeability to dextran FITC and, while had no effect on serum lipids, increased the blood levels of markers of liver injury and insulin resistance. DSS administration associated with low level inflammation of intestinal and mesenteric adipose tissues, caused liver histopathology features of steatohepatitis and severe atherosclerotic lesions in the aorta. These changes were prevented by VSL#3 intervention. Specifically, VSL#3 reversed insulin resistance, prevented development of histologic features of mesenteric adipose tissue inflammation, steatohepatitis and reduced the extent of aortic plaques. Conditioned media obtained from cultured probiotics caused the direct transactivation of peroxisome proliferator-activated receptor-γ, Farnesoid-X-receptors and vitamin D receptor. CONCLUSIONS Low grade intestinal inflammation drives a transition from steatosis to steatohepatitis and worsens the severity of atherosclerosis in a genetic model of dyslipidemia. VSL#3 intervention modulates the expression of nuclear receptors, corrects for insulin resistance in liver and adipose tissues and protects against development of steatohepatitis and atherosclerosis.
Collapse
Affiliation(s)
- Andrea Mencarelli
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
| | - Sabrina Cipriani
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
| | - Barbara Renga
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
| | - Angela Bruno
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
| | - Claudio D'Amore
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
| | | | - Stefano Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Facoltà di Medicina e Chirurgia, Perugia, Italy
- * E-mail:
| |
Collapse
|
40
|
Mariman R, Kremer B, van Erk M, Lagerweij T, Koning F, Nagelkerken L. Gene expression profiling identifies mechanisms of protection to recurrent trinitrobenzene sulfonic acid colitis mediated by probiotics. Inflamm Bowel Dis 2012; 18:1424-33. [PMID: 22162025 DOI: 10.1002/ibd.22849] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/08/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Host-microbiota interactions in the intestinal mucosa play a major role in intestinal immune homeostasis and control the threshold of local inflammation. The aim of this study was to evaluate the efficacy of probiotics in the recurrent trinitrobenzene sulfonic acid (TNBS)-induced colitis model and gain more insight into protective mechanisms. METHODS Moderate chronic inflammation of the colon was induced in BALB/c mice by repetitive intrarectal challenges with TNBS. Administration of probiotics started 2 weeks before colitis induction and was continued throughout colitis development. RESULTS Long-term administration of Lactobacillus plantarum NCIMB8826 or the probiotic mixture VSL#3 reduced intestinal inflammation induced by TNBS, evident from improved colon morphology and less influx of innate (CD11b(+) ) and adaptive (CD4(+) /CD8(+) ) immune cells in the intestinal mucosa and decreased proinflammatory serum cytokines (interferon-gamma [IFN-γ], interleukin [IL]-17, IL-1β, monocyte chemoattractant protein [MCP]-1) in probiotic-treated mice. Genomewide expression analysis of colonic tissues using microarrays revealed differences in expression of genes related to inflammation and immune processes between untreated and probiotic treated mice. Principal component analysis revealed that probiotic treatment resulted in a shift of gene expression profiles toward those of healthy controls. Effects of probiotics on colonic gene expression were most profound during active inflammation, in particular on gene clusters related to mast cells and antimicrobial peptides. The results were substantiated by suppression of chemokine gene expression. CONCLUSIONS Our data are in favor of a model in which probiotics downregulate expression of chemokines in the colon, thereby decreasing influx of inflammatory cells and rendering mice resistant to the induction of colitis.
Collapse
Affiliation(s)
- Rob Mariman
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
41
|
Caiozzi G, Wong BS, Ricketts ML. Dietary modification of metabolic pathways via nuclear hormone receptors. Cell Biochem Funct 2012; 30:531-51. [PMID: 23027406 DOI: 10.1002/cbf.2842] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 04/07/2012] [Accepted: 05/09/2012] [Indexed: 12/17/2022]
Abstract
Nuclear hormone receptors (NHRs), as ligand-dependent transcription factors, have emerged as important mediators in the control of whole body metabolism. Because of the promiscuous nature of several members of this superfamily that have been found to bind ligand with lower affinity than the classical steroid NHRs, they consequently display a broader ligand selectivity. This promiscuous nature has facilitated various bioactive dietary components being able to act as agonist ligands for certain members of the NHR superfamily. By binding to these NHRs, bioactive dietary components are able to mediate changes in various metabolic pathways, including, glucose, cholesterol and triglyceride homeostasis among others. This review will provide a general overview of the nuclear hormone receptors that have been shown to be activated by dietary components. The physiological consequences of such receptor activation by these dietary components will then be discussed in more detail.
Collapse
Affiliation(s)
- Gianella Caiozzi
- Department of Agriculture, Nutrition and Veterinary Sciences, University of Nevada Reno, Reno, NV 89557, USA
| | | | | |
Collapse
|
42
|
Ganesh BP, Richter JF, Blaut M, Loh G. Enterococcus faecium NCIMB 10415 does not protect interleukin-10 knock-out mice from chronic gut inflammation. Benef Microbes 2012; 3:43-50. [PMID: 22348908 DOI: 10.3920/bm2011.0050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Enterococcus faecium NCIMB 10415 reduces diarrhoea incidence and duration in animals and human study subjects. We tested whether the strain is also capable of reducing chronic gut inflammation and aimed to identify mechanisms that are involved in possible probiotic effects. To identify health-promoting mechanisms of the strain, we used interleukin-10-deficient mice that spontaneously develop gut inflammation and fed these mice a diet containing NCIMB 10415 for 3, 8 and 24 weeks, respectively. Control mice were fed a diet which was identically composed but did not contain the strain. After 3 weeks of intervention the experimental animals were less inflamed in the caecum than the control animals. This effect was not observed in the colon and there were no differences between experimental and control mice at any other time point. The application of the strain was associated with higher expression levels of interferon gamma and interferon gamma-induced protein 10 after 3 and 24 but not after 8 weeks of feeding. No differences between the animals were observed in intestinal barrier function or intestinal microbiota composition. However, we observed a low abundance of the mucin-degrading bacterium Akkermansia muciniphila in the mice that were fed NCIMB 10415 for 8 weeks. These low cell numbers were associated with a significantly lower caecal inflammation score and improved paracellular permeability as compared to the NCIMB-treated mice that were killed after 3 and 24 weeks of intervention. In conclusion, NCIMB 10415 is not capable of reducing gut inflammation in the IL-10-/- mouse model. The exact role of A. muciniphila and of a possible interaction between this bacterium, NCIMB 10415 and the host in gut inflammation requires further investigation.
Collapse
Affiliation(s)
- B P Ganesh
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Gastrointestinal Microbiology, Nuthetal, Germany.
| | | | | | | |
Collapse
|
43
|
Lactobacillus paracasei and Lactobacillus plantarum strains downregulate proinflammatory genes in an ex vivo system of cultured human colonic mucosa. GENES AND NUTRITION 2012; 8:165-80. [PMID: 22669626 DOI: 10.1007/s12263-012-0301-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/18/2012] [Indexed: 02/07/2023]
Abstract
Significant health benefits have been demonstrated for certain probiotic strains through intervention studies; however, there is a shortage of experimental evidence relative to the mechanisms of action. Here, noninvasive experimental procedure based on a colon organ culture system has been used that, in contrast to most experimental in vitro models reported, can preserve natural immunohistochemical features of the human mucosa. This system has been used to test whether commensal lactobacilli (Lactobacillus paracasei BL23, Lactobacillus plantarum 299v and L. plantarum 299v (A(-))) were able to hinder inflammation-like signals induced by phorbol 12-myristate 13-acetate (PMA)/ionomycin (IO). Whole genome microarrays have been applied to analyze expression differences, from which mRNA markers could be inferred to monitor the effect of putative probiotic strains under such conditions. Regarding the gene expression, PMA/IO treatment induced not only interleukin (IL)-2 and interferon gamma (IFN-γ), as expected, but also other relevant genes related to immune response and inflammation, such as IL-17A, chemokine (C-X-C motif) ligand (CXCL) 9 and CXCL11. The ex vivo culturing did not modify the pattern of expression of those genes or others related to inflammation. Interestingly, this study demonstrated that lactobacilli downregulated those genes and triggered a global change of the transcriptional profile that indicated a clear homeostasis restoring effect and a decrease in signals produced by activated T cells.
Collapse
|
44
|
Patterson AM, Delday MI, van Kuppevelt TH, Loh G, Blaut M, Haller D, Grant G, Kelly D. Expression of heparan sulfate proteoglycans in murine models of experimental colitis. Inflamm Bowel Dis 2012; 18:1112-26. [PMID: 21987406 DOI: 10.1002/ibd.21879] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 08/08/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND Heparan sulfate proteoglycans (HSPGs) are considered important in maintaining physiological homeostasis in many systems. Their expression is altered greatly in several pathophysiological conditions. Herein, we assess the expression and cellular localization of HSPGs in two murine models of human inflammatory bowel disease (IBD). METHODS Expression and localization of HSPGs, syndecans, and HS epitopes were examined in the colon of 129SvEv interleukin 10 knockout (IL10(-/-)), C3Bir IL10(-/-), and their genetic control (IL10(+/+)) counterparts (129SvEv; C3H/HeJ). mRNA expression of syndecans and heparan sulfate biosynthesis enzymes were evaluated by real-time polymerase chain reaction (PCR). Localization of HSPGs was determined by immunofluorescence. RESULTS mRNA for all syndecans was detected and expression in colonic tissues altered in IL10(-/-) mice. Syndecan-1 protein was expressed in the intestinal epithelium and on lamina propria cells of IL10(-/-) and control mice but was significantly reduced on the intestinal epithelial cells of IL10(-/-), mice particularly with severe colitis. Syndecan-2 was not detected, whereas syndecan-3 immunoreactivity was localized in the lamina propria but did not differ between control and IL10(-/-) mice. Syndecan-4 was present on epithelial cells of all mice but was significantly reduced in IL10(-/-) mice. Differences in the expression of HS epitopes between control and IL10(-/-) mice were also confirmed. CONCLUSIONS The study has revealed altered expression of syndecan-1 and -4 and HS epitopes in the gut of mice with an IBD-like gut disorder. The IL10(-/-) mouse is a useful model for further study of the functional role of HSPGs in chronic inflammation and in maintaining healthy gut barrier.
Collapse
Affiliation(s)
- A M Patterson
- Gut Immunology Group, Gut Health Division, Rowett Institute of Nutrition and Health, University of Aberdeen, Scotland, UK.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Arboleya S, González S, Salazar N, Ruas-Madiedo P, de los Reyes-Gavilán CG, Gueimonde M. Development of probiotic products for nutritional requirements of specific human populations. Eng Life Sci 2012. [DOI: 10.1002/elsc.201100129] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Sonia González
- Department of Functional Biology; University of Oviedo; Oviedo; Asturias; Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products; Instituto de Productos Lácteos de Asturias (IPLA-CSIC); Villaviciosa; Asturias; Spain
| |
Collapse
|
46
|
Chen YP, Hsiao PJ, Hong WS, Dai TY, Chen MJ. Lactobacillus kefiranofaciens M1 isolated from milk kefir grains ameliorates experimental colitis in vitro and in vivo. J Dairy Sci 2012; 95:63-74. [PMID: 22192184 DOI: 10.3168/jds.2011-4696] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/25/2011] [Indexed: 12/13/2022]
Abstract
Lactobacillus kefiranofaciens M1, isolated from and identified in Taiwanese milk kefir grain, has demonstrated immune-modulating activity. In the present study, we further investigated the effects of Lb. kefiranofaciens M1 on intestinal epithelial cells in vitro and on dextran sodium sulfate (DSS)-induced colitis in vivo. The possible mechanisms regarding the cytokine products and intestinal epithelial barrier restoration as well as the putative receptor for the protective effects of Lb. kefiranofaciens M1 were investigated. In vitro results indicated that Lb. kefiranofaciens M1 could strengthen the epithelial barrier function in vitro by increasing the transepithelial electrical resistance (TEER) and significantly upregulated the level of the chemokine CCL-20 at both the apical and basolateral sites. The in vivo effects of Lb. kefiranofaciens M1 on the regulation of intestinal physiology indicate that this strain could ameliorate DSS-induced colitis with a significant attenuation of the bleeding score and colon length shortening. Production of proinflammatory cytokines was decreased and that of the antiinflammatory cytokine IL-10 was increased in the DSS-treated mice given Lb. kefiranofaciens M1. The putative receptor for the protective effects of Lb. kefiranofaciens M1 was toll-like receptor 2 (TLR2), which was involved in probiotic-induced cytokine production in vitro and in attenuation of the bleeding score and colon length shortening in vivo. In this study, the kefir lactobacillus Lb. kefiranofaciens M1 clearly demonstrated an anticolitis effect. Based on these results, Lb. kefiranofaciens M1 has the potential to be applied in fermented dairy products as an alternative therapy for intestinal disorders.
Collapse
Affiliation(s)
- Y P Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
47
|
Kelly D, Delday MI, Mulder I. Microbes and microbial effector molecules in treatment of inflammatory disorders. Immunol Rev 2012; 245:27-44. [PMID: 22168412 DOI: 10.1111/j.1600-065x.2011.01079.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The healthy gut tolerates very large numbers of diverse bacterial species belonging mainly to the Bacteroidetes and Firmicutes phyla. These bacteria normally coexist peacefully with the gut and help maintain immune homeostasis and tolerance. The mechanisms promoting tolerance affect various cell populations, including the epithelial cells lining the gut, resident dendritic cells (DCs), and gut-homing T cells. Gut bacteria also influence multiple signaling pathways from Toll-like receptors to nuclear factor κB and regulate the functionality of DCs and T cells. Several bacterial species have been identified that promote T-cell differentiation, in particular T-helper 17 and T-regulatory cells. Insight into the molecular mechanisms by which bacteria mediate these effects will be very important in identifying new ways of treating intestinal and extra-intestinal immune-mediated diseases. These diseases are increasing dramatically in the human population and require new treatments. It may be possible in the future to identify specific bacterial species or strains that can correct for T-cell imbalances in the gut and promote immune homeostasis, both locally and systemically. In addition, new information describing microbial genomes affords the opportunity to mine for functional genes that may lead to new generation drugs relevant to a range of inflammatory disease conditions.
Collapse
Affiliation(s)
- Denise Kelly
- Gut Immunology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK.
| | | | | |
Collapse
|
48
|
Appleyard CB, Cruz ML, Isidro AA, Arthur JC, Jobin C, De Simone C. Pretreatment with the probiotic VSL#3 delays transition from inflammation to dysplasia in a rat model of colitis-associated cancer. Am J Physiol Gastrointest Liver Physiol 2011; 301:G1004-13. [PMID: 21903764 PMCID: PMC3233787 DOI: 10.1152/ajpgi.00167.2011] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Evidence supports involvement of microflora in the transition of chronic inflammation to neoplasia. We investigated the protective efficacy of the probiotic VSL#3 in a model of colitis-associated colorectal cancer. Chronic colitis was induced in Sprague-Dawley rats by administration of trinitrobenzene sulfonic acid (TNBS), followed 6 wk later by systemic reactivation. To induce colitis-associated dysplasia and cancer, the animals received TNBS (intravenously) twice a week for 10 wk. One group received VSL#3 in drinking water from 1 wk before colitis induction until death. The colons were examined for damage and presence of dysplasia or cancer. Samples were analyzed for cell proliferation and apoptosis, vitamin D receptor (VDR) expression, angiogenic factors, and presence of alkaline sphingomyelinase or phosphatase. Microbial community composition was evaluated by terminal restriction fragment-length polymorphism analysis of the bacterial 16S rRNA gene. None of the probiotic-treated animals developed carcinoma, and no high-grade dysplasia was found in either the proximal or mid colon. In contrast, 29% of the animals in the control group developed carcinoma in one or more regions of the colon. VSL#3-treated animals had significantly less damage than the vehicle treated-controls in all areas of the colon, and this correlated with decreased richness and diversity of the mucosally adherent microbiota. Treatment with the probiotic increased the antiangiogenic factor angiostatin, VDR expression, and alkaline sphingomyelinase. We concluded that pretreatment with the probiotic VSL#3 can attenuate various inflammatory-associated parameters, delaying transition to dysplasia and cancer, thus offering its potential therapeutic use in patients with long-standing colitis.
Collapse
Affiliation(s)
- Caroline B. Appleyard
- 1Department of Physiology and Pharmacology, Ponce School of Medicine, Ponce, Puerto Rico;
| | - Myrella L. Cruz
- 1Department of Physiology and Pharmacology, Ponce School of Medicine, Ponce, Puerto Rico;
| | - Angel A. Isidro
- 1Department of Physiology and Pharmacology, Ponce School of Medicine, Ponce, Puerto Rico;
| | - Janelle C. Arthur
- 2Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Christian Jobin
- 2Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - Claudio De Simone
- 3Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
49
|
Zakostelska Z, Kverka M, Klimesova K, Rossmann P, Mrazek J, Kopecny J, Hornova M, Srutkova D, Hudcovic T, Ridl J, Tlaskalova-Hogenova H. Lysate of probiotic Lactobacillus casei DN-114 001 ameliorates colitis by strengthening the gut barrier function and changing the gut microenvironment. PLoS One 2011; 6:e27961. [PMID: 22132181 PMCID: PMC3222668 DOI: 10.1371/journal.pone.0027961] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 10/28/2011] [Indexed: 12/12/2022] Open
Abstract
Background Probiotic bacteria can be used for the prevention and treatment of human inflammatory diseases including inflammatory bowel diseases (IBD). However, the nature of active components and exact mechanisms of this beneficial effects have not been fully elucidated. Our aim was to investigate if lysate of probiotic bacterium L. casei DN-114 001 (Lc) could decrease the severity of intestinal inflammation in a murine model of IBD. Methodology/Principal Findings The preventive effect of oral administration of Lc significantly reduces the severity of acute dextran sulfate sodium (DSS) colitis in BALB/c but not in SCID mice. In order to analyze how this beneficial effect interferes with well-known phases of intestinal inflammation pathogenesis in vivo and in vitro, we evaluated intestinal permeability using the FITC-labeled dextran method and analysed tight junction proteins expression by immunofluorescence and PCR. We also measured CD4+FoxP3+ regulatory T cells proportion by FACS analysis, microbiota composition by pyrosequencing, and local cytokine production by ELISA. Lc leads to a significant protection against increased intestinal permeability and barrier dysfunction shown by preserved ZO-1 expression. We found that the Lc treatment increases the numbers of CD4+FoxP3+ regulatory T cells in mesenteric lymph nodes (MLN), decreases production of pro-inflammatory cytokines TNF-α and IFN-γ, and anti-inflammatory IL-10 in Peyer's patches and large intestine, and changes the gut microbiota composition. Moreover, Lc treatment prevents lipopolysaccharide-induced TNF-α expression in RAW 264.7 cell line by down-regulating the NF-κB signaling pathway. Conclusion/Significance Our study provided evidence that even non-living probiotic bacteria can prevent the development of severe forms of intestinal inflammation by strengthening the integrity of intestinal barrier and modulation of gut microenvironment.
Collapse
Affiliation(s)
- Zuzana Zakostelska
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Miloslav Kverka
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| | - Klara Klimesova
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Rossmann
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jakub Mrazek
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jan Kopecny
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Michaela Hornova
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Dagmar Srutkova
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Tomas Hudcovic
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Novy Hradek, Czech Republic
| | - Jakub Ridl
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | |
Collapse
|
50
|
Shores DR, Binion DG, Freeman BA, Baker PR. New insights into the role of fatty acids in the pathogenesis and resolution of inflammatory bowel disease. Inflamm Bowel Dis 2011; 17:2192-204. [PMID: 21910181 PMCID: PMC4100336 DOI: 10.1002/ibd.21560] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 10/05/2010] [Indexed: 12/12/2022]
Abstract
Dietary and endogenously modified lipids modulate inflammation by functioning as intra- and intercellular signaling molecules. Proinflammatory lipid mediators such as the eicosanoids compete against the signaling actions of newly discovered modified fatty acids that act to resolve inflammation. In inflammatory bowel disease, multiple aberrancies in lipid metabolism have been discovered, which shed further light on the pathogenesis of intestinal inflammation. Mechanisms by which lipids modulate inflammation, abnormalities of lipid metabolism in the setting of inflammatory bowel disease, and potential therapeutic application of lipid derivatives in this setting are discussed.
Collapse
Affiliation(s)
- Darla R. Shores
- Division of Pediatric Gastroenterology, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - David G. Binion
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Bruce A. Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Paul R.S. Baker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|