1
|
Hong JB, Chen YX, Su ZY, Chen XY, Lai YN, Yang JH. Causal association of juvenile idiopathic arthritis or JIA-associated uveitis and gut microbiota: a bidirectional two-sample Mendelian randomisation study. Front Immunol 2024; 15:1356414. [PMID: 39114654 PMCID: PMC11303189 DOI: 10.3389/fimmu.2024.1356414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Background The gut microbiota significantly influences the onset and progression of juvenile idiopathic arthritis (JIA) and associated uveitis (JIAU); however, the causality remains unclear. This study aims to establish a causal link between gut microbiota and JIA or JIAU. Methods Using publicly available genome-wide association studies (GAWS) summary data, we conducted a two-sample Mendelian randomisation (MR) analysis employing various methods, namely inverse variance weighted (IVW), simple mode, weighted mode, weighted median and MR-Egger regression methods, to assess the causal association between JIA or JIAU and gut microbiota. Sensitivity analyses, including Cochrane's Q test, MR-Egger intercept test, leave-one-out analysis and MR-PRESSO, were performed to evaluate the robustness of the MR results. Subsequently, reverse MR analysis was conducted to determine causality between gene-predicted gut microbiota abundance and JIA or JIAU. Results The MR analysis revealed a causal association between gut microbiota abundance variations and JIA or JIAU risk. Specifically, the increased abundance of genus Ruminococcaceae UCG013 (OR: 0.055, 95%CI: 0.006-0.103, p = 0.026) and genus Ruminococcaceae UCG003 (β: 0.06, 95%CI: 0.003-0.117, p = 0.041) correlated with an increased risk of JIA, while genus Lachnospiraceae UCG001 (OR: 0.833, 95%CI: 0.699~0.993, p = 0.042) was associated with a reduced risk of JIA, among others. Sensitivity analysis confirmed MR analysis robustness. Conclusions This study provides substantial evidence supporting a causal association between genetically predicted gut microbiota and JIA or JIAU. It highlights the significant role of intestinal flora in JIA or JIAU development, suggesting their potential as novel biomarkers for diagnosis and prevention. These findings offer valuable insights to mitigate the impact of JIA or JIAU.
Collapse
Affiliation(s)
- Jun-bin Hong
- Department of Pediatrics, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue-xuan Chen
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Zhi-ying Su
- Department of Pediatrics, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin-ying Chen
- Department of Pediatrics, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan-ni Lai
- School of Medicine and Health, Shunde Polytechnic, Foshan, China
| | - Jing-hua Yang
- Department of Pediatrics, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- Xiaorong Luo’s National Renowned Expert Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Nagarajan G, Govindan R, Poomarimuthu M, Andiappan R, Elango S, Maruthamuthu S, Mariakuttikan J, Kadiam S. The microbiome and rheumatic heart disease: current knowledge and future perspectives. Acta Cardiol 2023:1-9. [PMID: 37171266 DOI: 10.1080/00015385.2023.2207933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Rheumatic heart disease (RHD) is a cardiovascular disease caused by an autoimmune response to group A Streptococcus (GAS) infection resulting in the damage of heart valves. RHD is the most commonly acquired heart disease among children and young adults with a global burden of over 40 million cases accounting for 306,000 deaths annually. Inflammation in the heart valves caused due to molecular mimicry between the GAS antigens and host cardiac proteins is facilitated by cytokines, cross-reactive antibodies and CD4+ T cells. The complex interaction between genetic and environmental factors linked with erratic events leads to the loss of immunological tolerance and autoimmunity in RHD. Despite extensive research on the etiopathogenesis of RHD, the precise mechanism underpinning the initiation of acute rheumatic fever (ARF) to the progression of RHD still remains elusive. Mounting evidences support the contribution of the human microbiome in the development of several immune-mediated diseases including rheumatoid arthritis, juvenile idiopathic arthritis, Kawasaki disease, inflammatory bowel disease and type 1 diabetes. The microbiome and their metabolites could play a crucial role in the integrity of the epithelial barrier, development of the immune system, inflammation and differentiation of T cell subsets. Consequently, microbiome dysbiosis might result in autoimmunity by molecular mimicry, epitope spreading and bystander activation. This review discusses various aspects of the interaction between the microbiome and the immune system in order to reveal causative links relating dysbiosis and autoimmune diseases with special emphasis on RHD.
Collapse
Affiliation(s)
- Gunavathy Nagarajan
- Department of Immunology, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Ramajayam Govindan
- Multidisciplinary Research Unit, Madurai Medical College, Madurai, India
| | | | - Rathinavel Andiappan
- Department of Cardio Vascular Thoracic Surgery, Madurai Medical College & Government Rajaji Hospital, Madurai, India
| | - Sivakumar Elango
- Institute of Child Health and Research Centre, Madurai Medical College & Government Rajaji Hospital, Madurai, India
| | - Stalinraja Maruthamuthu
- Department of Surgery, Immunogenetics and Transplantation Laboratory, University of California, San Francisco, CA, USA
| | | | - Sony Kadiam
- Department of Immunology, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| |
Collapse
|
3
|
Martinović A, Chittaro M, Mora D, Arioli S. The Ability of Streptococcus thermophilus BT01 to Modulate Urease Activity in Healthy Subjects' Fecal Samples Depends on the Biomass Production Process. Mol Nutr Food Res 2023; 67:e2200529. [PMID: 36708131 DOI: 10.1002/mnfr.202200529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/10/2023] [Indexed: 01/29/2023]
Abstract
SCOPE This study evaluates how manufacturing conditions of probiotic biomass production, using two different cryoprotectants, Cryo-A and Cryo-B, can affect Streptococcus thermophilus BT01 in vivo gastrointestinal tract survival and its ability to modulate the level of urease activity in fecal samples of healthy subjects. METHODS AND RESULTS A randomized controlled cross-over study is carried out on 20 adult healthy subjects to evaluate total and viable loads, persistence of S. thermophilus BT01, and urease activity in fecal samples. Strain-specific quantification by using developed culture-based method and molecular qPCR tool allows to quantify viable S. thermophilus BT01 strain in 90% of the subjects. The quantification of both total DNA and recovered viable S. thermophilus BT01 in fecal samples does not reveal significant differences between Cryo-A or Cryo-B treated biomass. However, the administration of S. thermophilus BT01 produced with Cryo-A results in a decreased urease activity in fecal samples compared to Cryo-B protected cells. CONCLUSION This study i) highlights how the manufacturing conditions can play a role in influencing the probiotic functionality in vivo and ii) represents the first evidence that links S. thermophilus to a specific probiotic mechanism, the reduction of urease activity in fecal samples.
Collapse
Affiliation(s)
- Anđela Martinović
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, Milan, 20122, Italy
| | - Marco Chittaro
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, Milan, 20122, Italy
| | - Diego Mora
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, Milan, 20122, Italy
| | - Stefania Arioli
- Department of Food, Environmental, and Nutritional Sciences (DeFENS), University of Milan, Milan, 20122, Italy
| |
Collapse
|
4
|
Cruz N, Abernathy GA, Dichosa AEK, Kumar A. The Age of Next-Generation Therapeutic-Microbe Discovery: Exploiting Microbe-Microbe and Host-Microbe Interactions for Disease Prevention. Infect Immun 2022; 90:e0058921. [PMID: 35384688 PMCID: PMC9119102 DOI: 10.1128/iai.00589-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Humans are considered "superorganisms," harboring a diverse microbial collective that outnumbers human cells 10 to 1. Complex and gravely understudied host- and microbe-microbe interactions-the product of millions of years of host-microbe coevolution-govern the superorganism in almost every aspect of life functions and overall well-being. Abruptly disrupting these interactions via extrinsic factors has undesirable consequences for the host. On the other hand, supplementing commensal or beneficial microbes may mitigate perturbed interactions or enhance the interactive relationships that ultimately benefit all parties. Hence, immense efforts have focused on dissecting the innumerable host- and microbe-microbe relationships to characterize if a "positive" or "negative" interaction is at play and to exploit such behavior for broader implications. For example, microbiome research has worked to identify and isolate naturally antipathogenic microbes that may offer therapeutic potential either in a direct, one-on-one application or by leveraging its unique metabolic properties. However, the discovery and isolation of such desired therapeutic microbes from complex microbiota have proven challenging. Currently, there is no conventional technique to universally and functionally screen for these microbes. With this said, we first describe in this review the historical (probiotics) and current (fecal microbiota or defined consortia) perspectives on therapeutic microbes, present the discoveries of therapeutic microbes through exploiting microbe-microbe and host-microbe interactions, and detail our team's efforts in discovering therapeutic microbes via our novel microbiome screening platform. We conclude this minireview by briefly discussing challenges and possible solutions with therapeutic microbes' applications and paths ahead for discovery.
Collapse
Affiliation(s)
- Nathan Cruz
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - George A. Abernathy
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Armand E. K. Dichosa
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Anand Kumar
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| |
Collapse
|
5
|
Frid P, Baraniya D, Halbig J, Rypdal V, Songstad NT, Rosèn A, Berstad JR, Flatø B, Alakwaa F, Gil EG, Cetrelli L, Chen T, Al-Hebshi NN, Nordal E, Al-Haroni M. Salivary Oral Microbiome of Children With Juvenile Idiopathic Arthritis: A Norwegian Cross-Sectional Study. Front Cell Infect Microbiol 2020; 10:602239. [PMID: 33251163 PMCID: PMC7672027 DOI: 10.3389/fcimb.2020.602239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background The oral microbiota has been connected to the pathogenesis of rheumatoid arthritis through activation of mucosal immunity. The objective of this study was to characterize the salivary oral microbiome associated with juvenile idiopathic arthritis (JIA), and correlate it with the disease activity including gingival inflammation. Methods Fifty-nine patients with JIA (mean age, 12.6 ± 2.7 years) and 34 healthy controls (HC; mean age 12.3 ± 3.0 years) were consecutively recruited in this Norwegian cross-sectional study. Information about demographics, disease activity, medication history, frequency of tooth brushing and a modified version of the gingival bleeding index (GBI) and the simplified oral hygiene index (OHI-S) was obtained. Microbiome profiling of saliva samples was performed by sequencing of the V1-V3 region of the 16S rRNA gene, coupled with a species-level taxonomy assignment algorithm; QIIME, LEfSe and R-package for Spearman correlation matrix were used for downstream analysis. Results There were no significant differences between JIA and HC in alpha- and beta-diversity. However, differential abundance analysis revealed several taxa to be associated with JIA: TM7-G1, Solobacterium and Mogibacterium at the genus level; and Leptotrichia oral taxon 417, TM7-G1 oral taxon 352 and Capnocytophaga oral taxon 864 among others, at the species level. Haemophilus species, Leptotrichia oral taxon 223, and Bacillus subtilis, were associated with healthy controls. Gemella morbillorum, Leptotrichia sp. oral taxon 498 and Alloprevotella oral taxon 914 correlated positively with the composite juvenile arthritis 10-joint disease activity score (JADAS10), while Campylobacter oral taxon 44 among others, correlated with the number of active joints. Of all microbial markers identified, only Bacillus subtilis and Campylobacter oral taxon 44 maintained false discovery rate (FDR) < 0.1. Conclusions In this exploratory study of salivary oral microbiome we found similar alpha- and beta-diversity among children with JIA and healthy. Several taxa associated with chronic inflammation were found to be associated with JIA and disease activity, which warrants further investigation.
Collapse
Affiliation(s)
- Paula Frid
- Department of ENT, Division of Oral and Maxillofacial Surgery, University Hospital North Norway, Tromsø, Norway.,Public Dental Service Competence Centre of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway
| | - Divyashri Baraniya
- Oral Microbiome Laboratory, Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Josefine Halbig
- Public Dental Service Competence Centre of North Norway, Tromsø, Norway.,Department of Clinical Dentistry, UiT the Arctic University of Norway, Tromsø, Norway
| | - Veronika Rypdal
- Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway.,Department of Pediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Nils Thomas Songstad
- Department of Pediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Annika Rosèn
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway.,Department of Oral and Maxillofacial Surgery, Haukeland University Hospital, Bergen, Norway
| | - Johanna Rykke Berstad
- Department of ENT, Division of Oral and Maxillofacial Surgery, Oslo University Hospital, Oslo, Norway
| | - Berit Flatø
- Department of Rheumatology and Infectious Diseases, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Rheumatology, Oslo University Hospital, Oslo, Norway
| | - Fadhl Alakwaa
- Department of Computational Medicine and Bioinformatics, University Michigan, Ann Arbor, MI, United States
| | | | - Lena Cetrelli
- Center of Oral Health Services and Research (TkMidt), Trondheim, Norway
| | - Tsute Chen
- Department of Microbiology, Forsyth Institute, Cambridge, MA, United States
| | - Nezar Noor Al-Hebshi
- Oral Microbiome Laboratory, Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Ellen Nordal
- Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway.,Department of Pediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Mohammed Al-Haroni
- Department of Clinical Dentistry, UiT the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
van Loosdregt J, van Wijk F, Prakken B, Vastert B. Update on research and clinical translation on specific clinical areas from biology to bedside: Unpacking the mysteries of juvenile idiopathic arthritis pathogenesis. Best Pract Res Clin Rheumatol 2018; 31:460-475. [PMID: 29773267 DOI: 10.1016/j.berh.2018.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 02/08/2023]
Abstract
In the past decades, we have gained important insights into the mechanisms of disease and therapy underlying chronic inflammation in juvenile idiopathic arthritis (JIA). These insights have resulted in several game-changing therapeutic modalities for many patients. However, additional progress still has to be made with regard to efficacy, cost reduction, minimization of side effects, and dose-tapering and stop strategies of maintenance drugs. Moreover, to really transform the current therapeutic strategies into personalized medicine, we need validated biomarkers to translate increased insights into clinical practice. In this article, we describe recent developments in JIA research and outline how clinical innovations need to go hand in hand with basic discoveries to really effect care for patients. Facilitating the transition from bench to bedside is crucial for addressing the major current challenges in JIA management. When successful, it will set new standards for a safe, targeted, and personalized medicine in JIA.
Collapse
Affiliation(s)
- Jorg van Loosdregt
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Femke van Wijk
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Berent Prakken
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands
| | - Bas Vastert
- Department of Pediatric Immunology & Rheumatology, Laboratory for Translational Medicine, University Medical Centre Utrecht, University of Utrecht, Lundlaan 6, P.O. Box 85090, 3584 EA/3508 AB, Utrecht, The Netherlands.
| |
Collapse
|
7
|
Khanna S, Jaiswal KS, Gupta B. Managing Rheumatoid Arthritis with Dietary Interventions. Front Nutr 2017; 4:52. [PMID: 29167795 PMCID: PMC5682732 DOI: 10.3389/fnut.2017.00052] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Self-help by means of dietary interventions can help in management of various disorders including rheumatoid arthritis (RA), a debilitating autoimmune disease. Dietary interventions necessitate a widespread appeal for both patients as well as clinicians due to factors including affordability, accessibility, and presence of scientific evidences that demonstrate substantial benefits in reducing disease symptoms such as pain, joint stiffness, swelling, tenderness and associated disability with disease progression. However, there is still an uncertainty among the community about the therapeutic benefits of dietary manipulations for RA. In the present review, we provide an account of different diets and their possible molecular mechanism of actions inducing observed therapeutic benefits for remission and management of RA. We further indicate food that can be a potential aggravating factor for the disease or may help in symptomatic relief. We thereafter summarize and thereby discuss various diets and food which help in reducing levels of inflammatory cytokines in RA patients that may play an effective role in management of RA following proper patient awareness. We thus would like to promote diet management as a tool that can both supplement and complement present treatment strategies for a better patient health and recovery.
Collapse
Affiliation(s)
- Shweta Khanna
- Disease Biology Laboratory, School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Kumar Sagar Jaiswal
- Disease Biology Laboratory, School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Bhawna Gupta
- Disease Biology Laboratory, School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
8
|
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with progressive joint disorder. The complex interplay of genetic and environmental influences is important for the development of the disease. A growing body of evidence has shed light on the association of dysbiosis of gut microbiota with RA. Certain gut microbial strains have been shown to inhibit or attenuate immune responses in RA experimental models, suggesting that specific species among intestinal commensal bacteria may play either a pathogenic or a protective role in the development of RA. Oral intake of probiotics/prebiotics can therefore represent a therapeutic approach for RA treatment. However, the relevant scientific work has only just begun, and the available data in this field remain limited. Fortunately, utilization of new sequencing technologies allows expanded research on the association of intestinal bacterial flora and human diseases to be attempted. In this review, we summarize the role of gut microbiota in RA progression and address how specific bacterial strains regulate the immune response in disease process. Probiotics/prebiotics in the treatment of RA is also discussed.
Collapse
Affiliation(s)
- Y Kang
- Medical Faculty, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - Y Cai
- Medical Faculty, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - X Zhang
- Medical Faculty, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - X Kong
- Medical Faculty, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China
| | - J Su
- Medical Faculty, Kunming University of Science and Technology, 650500, Kunming, Yunnan, China.
| |
Collapse
|
9
|
Verwoerd A, Ter Haar NM, de Roock S, Vastert SJ, Bogaert D. The human microbiome and juvenile idiopathic arthritis. Pediatr Rheumatol Online J 2016; 14:55. [PMID: 27650128 PMCID: PMC5028952 DOI: 10.1186/s12969-016-0114-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/15/2016] [Indexed: 01/05/2023] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common rheumatic disease in childhood. The pathogenesis of JIA is thought to be the result of a combination of host genetic and environmental triggers. However, the precise factors that determine one's susceptibility to JIA remain to be unravelled. The microbiome has received increasing attention as a potential contributing factor to the development of a wide array of immune-mediated diseases, including inflammatory bowel disease, type 1 diabetes and rheumatoid arthritis. Also in JIA, there is accumulating evidence that the composition of the microbiome is different from healthy individuals. A growing body of evidence indeed suggests that, among others, the microbiome may influence the development of the immune system, the integrity of the intestinal mucosal barrier, and the differentiation of T cell subsets. In turn, this might lead to dysregulation of the immune system, thereby possibly playing a role in the development of JIA. The potential to manipulate the microbiome, for example by faecal microbial transplantation, might then offer perspectives for future therapeutic interventions. Before we can think of such interventions, we need to first obtain a deeper understanding of the cause and effect relationship between JIA and the microbiome. In this review, we discuss the existing evidence for the involvement of the microbiome in JIA pathogenesis and explore the potential mechanisms through which the microbiome may influence the development of autoimmunity in general and JIA specifically.
Collapse
Affiliation(s)
- Anouk Verwoerd
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Nienke M. Ter Haar
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Sytze de Roock
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Sebastiaan J. Vastert
- Laboratory of Translational Immunology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands ,Department of Paediatric Rheumatology, Wilhelmina Children’s Hospital, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Debby Bogaert
- Department of Paediatric Infectious Diseases, Wilhelmina Children's Hospital, Lundlaan 6, 3584 EA, Utrecht, The Netherlands.
| |
Collapse
|
10
|
Wang P, Tao JH, Pan HF. Probiotic bacteria: a viable adjuvant therapy for relieving symptoms of rheumatoid arthritis. Inflammopharmacology 2016; 24:189-196. [PMID: 27581587 DOI: 10.1007/s10787-016-0277-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/23/2016] [Indexed: 01/05/2023]
Abstract
The burgeoning use of probiotics has proliferated during the past two decades. However, the effect of probiotic administration for either the prevention or treatment of rheumatoid arthritis (RA) has been investigated in a limited number of studies. Randomized controlled clinical trials have provided evidences that specific probiotics supplementation exhibit anti-inflammatory effects, help to increase daily activities and alleviate symptoms in patients with RA. Therefore, using probiotic bacteria as an adjuvant therapy may be considered as a promising treatment option for RA. This review summarizes the available data about the therapeutic and preventive effect of probiotics in RA, together with probiotic supplement as a possible therapy in clinical treatment.
Collapse
Affiliation(s)
- Peng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Laboratory of Population Health & Major Disease Screening and Diagnosis, Hefei, Anhui, China
| | - Jin-Hui Tao
- Department of Rheumatology, Anhui Provincial Hospital, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Laboratory of Population Health & Major Disease Screening and Diagnosis, Hefei, Anhui, China.
| |
Collapse
|
11
|
Tejesvi MV, Arvonen M, Kangas SM, Keskitalo PL, Pirttilä AM, Karttunen TJ, Vähäsalo P. Faecal microbiome in new-onset juvenile idiopathic arthritis. Eur J Clin Microbiol Infect Dis 2015; 35:363-70. [PMID: 26718942 DOI: 10.1007/s10096-015-2548-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023]
Abstract
Alterations in the intestinal microbial flora have been linked with autoimmune diseases. Our objective was to analyse the composition of the faecal microbiome of children with new-onset juvenile idiopathic arthritis (JIA) compared to healthy controls, and to identify specific gut bacteria associated with JIA. Stool samples from patients were taken at the time of diagnosis of JIA. The microbiome profiles of samples of 30 children with JIA (mean age 6.2 years, 22 girls) were analysed with 16S region-based sequencing profiling and compared to the stool samples of healthy controls (n = 27, mean age 5.4 years, 18 girls). The proportion of bacteria belonging to the phylum Firmicutes was significantly lower in children with JIA [21 % (95 % confident interval [CI]: 17-25 %)] compared to controls [33 % (95 % CI: 26-41 %), p = 0.009]. Bacteria belonging to Bacteroidetes were significantly more abundant in JIA [78 % (95 % CI: 74-82 %)] than in control samples [65 % (95 % CI: 57-73 %), p = 0.008]. Shared operational taxonomic units (OTUs) between the groups revealed that genera Actinobacteria and Fusobacteria were present only in JIA patients and Lentisphaerae only in controls. In summary, faecal flora in JIA is characterised by a low level of Firmicutes and an abundance of Bacteroidetes, resembling the aberration reported in type 1 diabetes. We suggest that alterations in the intestinal microbial flora may challenge the mucosal immune system of genetically susceptible subjects predisposing to local proinflammatory cascades, thus contributing to the development of JIA.
Collapse
Affiliation(s)
- M V Tejesvi
- Genetics and Physiology, Faculty of Science, University of Oulu, Oulu, Finland
| | - M Arvonen
- PEDEGO Research Unit, University of Oulu, Oulu, Finland. .,Department of Paediatrics and Adolescents, Oulu University Hospital, Oulu, Finland. .,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland. .,Department of Paediatrics, Kuopio University Hospital, Kuopio, Finland.
| | - S M Kangas
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Department of Paediatrics and Adolescents, Oulu University Hospital, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Medical Microbiology and Immunology, Research Group of Biomedicine, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - P L Keskitalo
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Department of Paediatrics and Adolescents, Oulu University Hospital, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - A M Pirttilä
- Genetics and Physiology, Faculty of Science, University of Oulu, Oulu, Finland
| | - T J Karttunen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Department of Pathology, Oulu University Hospital, Oulu, Finland
| | - P Vähäsalo
- PEDEGO Research Unit, University of Oulu, Oulu, Finland.,Department of Paediatrics and Adolescents, Oulu University Hospital, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
12
|
Kabeerdoss J, Sandhya P, Danda D. Gut inflammation and microbiome in spondyloarthritis. Rheumatol Int 2015; 36:457-68. [PMID: 26719306 DOI: 10.1007/s00296-015-3414-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022]
Abstract
Spondyloarthritis (SpA) is chronic inflammatory disease involving joints and the spine. Bowel inflammation is common in SpA, which may be classified as acute or chronic. Chronic gut inflammation is most common in SpA patients with axial involvement as compared to those presenting with peripheral involvement alone. The pathogenesis of gut inflammation in SpA could be explained by two factors-over-activation of immunological cells and altered gut microbiome. This is exemplified by SpA animal models, namely HLA-B27-expressing transgenic animals and SKG mice models. Immunological mechanisms include homing of activated T cells from gut into synovium, excess pro-inflammatory cytokines secretion by immune cells such as IL-23 and genetic variations in immunological genes. The evidence for role of gut microbiome in SpA is gradually emerging. Recently, metagenomic study of gut microbiome by sequencing of microbial nucleic acids has enabled identification of new microbial taxa and their functions in gut of patients with SpA. In SpA, the gut microbiome could emerge as diagnostic and prognostic marker of disease. Modulation of gut microbiome is slated to have therapeutic potential as well.
Collapse
Affiliation(s)
- Jayakanthan Kabeerdoss
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Pulukool Sandhya
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India.
| |
Collapse
|
13
|
Vaghef-Mehrabany E, Homayouni-Rad A, Alipour B, Sharif SK, Vaghef-Mehrabany L, Alipour-Ajiry S. Effects of Probiotic Supplementation on Oxidative Stress Indices in Women with Rheumatoid Arthritis: A Randomized Double-Blind Clinical Trial. J Am Coll Nutr 2015; 35:291-9. [DOI: 10.1080/07315724.2014.959208] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
Arvonen M, Virta LJ, Pokka T, Kröger L, Vähäsalo P. Repeated exposure to antibiotics in infancy: a predisposing factor for juvenile idiopathic arthritis or a sign of this group's greater susceptibility to infections? J Rheumatol 2014; 42:521-6. [PMID: 25320218 DOI: 10.3899/jrheum.140348] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Previous exposure to antibiotics has been associated with the pathogenesis of several autoimmune diseases. Our objective was to explore whether childhood exposure to antibiotics would be associated with the risk of developing juvenile idiopathic arthritis (JIA). METHODS The material was collected from national registers containing all children born in 2000-2010 in Finland and diagnosed with JIA by the end of December 2012 (n = 1298) and appropriate controls (n = 5179) matched for age, sex, and place of birth. All purchases of antibiotics were collected from birth until the index date (i.e., the date of special reimbursement for JIA medications). A conditional logistic regression was performed to evaluate the association between the exposure to antibiotics and the risk of JIA. RESULTS The risk of JIA increased with the number of antibiotic purchases from birth to the index date: for ≥ 1 purchases versus none, OR 1.6, 95% CI 1.3-1.9 with an upward trend in OR (p < 0.001). Antibiotic groups lincosamides and cephalosporins showed the strongest association with JIA (OR 6.6, 95% CI 3.7-11.7, and OR 1.6, 95% CI 1.4-1.8, respectively). Overall exposure to antibiotics before 2 years of age was associated with an increased risk of JIA (OR 1.4, 95% CI 1.2-1.6), with the trend test of OR (p < 0.001). CONCLUSION Previous early and repeated exposure to antibiotics may predispose individuals to develop JIA. Alternatively, the apparent association may reflect shared susceptibility to infections and JIA.
Collapse
Affiliation(s)
- Miika Arvonen
- From the Department of Pediatrics, Kuopio University Hospital, Kuopio, and University of Eastern Finland, Kuopio; Research Department, Social Insurance Institution, Turku; Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland.M. Arvonen, MD, PhD, Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, and Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, University of Oulu; L.J. Virta, MD, PhD, Research Department, Social Insurance Institution; T. Pokka, MSc, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu; L. Kröger, MD, PhD, Department of Pediatrics, Kuopio University Hospital, and University of Eastern Finland; P. Vähäsalo, MD, PhD, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu.
| | - Lauri J Virta
- From the Department of Pediatrics, Kuopio University Hospital, Kuopio, and University of Eastern Finland, Kuopio; Research Department, Social Insurance Institution, Turku; Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland.M. Arvonen, MD, PhD, Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, and Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, University of Oulu; L.J. Virta, MD, PhD, Research Department, Social Insurance Institution; T. Pokka, MSc, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu; L. Kröger, MD, PhD, Department of Pediatrics, Kuopio University Hospital, and University of Eastern Finland; P. Vähäsalo, MD, PhD, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu
| | - Tytti Pokka
- From the Department of Pediatrics, Kuopio University Hospital, Kuopio, and University of Eastern Finland, Kuopio; Research Department, Social Insurance Institution, Turku; Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland.M. Arvonen, MD, PhD, Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, and Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, University of Oulu; L.J. Virta, MD, PhD, Research Department, Social Insurance Institution; T. Pokka, MSc, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu; L. Kröger, MD, PhD, Department of Pediatrics, Kuopio University Hospital, and University of Eastern Finland; P. Vähäsalo, MD, PhD, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu
| | - Liisa Kröger
- From the Department of Pediatrics, Kuopio University Hospital, Kuopio, and University of Eastern Finland, Kuopio; Research Department, Social Insurance Institution, Turku; Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland.M. Arvonen, MD, PhD, Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, and Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, University of Oulu; L.J. Virta, MD, PhD, Research Department, Social Insurance Institution; T. Pokka, MSc, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu; L. Kröger, MD, PhD, Department of Pediatrics, Kuopio University Hospital, and University of Eastern Finland; P. Vähäsalo, MD, PhD, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu
| | - Paula Vähäsalo
- From the Department of Pediatrics, Kuopio University Hospital, Kuopio, and University of Eastern Finland, Kuopio; Research Department, Social Insurance Institution, Turku; Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland.M. Arvonen, MD, PhD, Department of Pediatrics, Kuopio University Hospital, University of Eastern Finland, and Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, University of Oulu; L.J. Virta, MD, PhD, Research Department, Social Insurance Institution; T. Pokka, MSc, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu; L. Kröger, MD, PhD, Department of Pediatrics, Kuopio University Hospital, and University of Eastern Finland; P. Vähäsalo, MD, PhD, Medical Research Center Oulu, Department of Pediatrics, Oulu University Hospital, and University of Oulu
| |
Collapse
|
15
|
Alipour B, Homayouni-Rad A, Vaghef-Mehrabany E, Sharif SK, Vaghef-Mehrabany L, Asghari-Jafarabadi M, Nakhjavani MR, Mohtadi-Nia J. Effects of Lactobacillus casei supplementation on disease activity and inflammatory cytokines in rheumatoid arthritis patients: a randomized double-blind clinical trial. Int J Rheum Dis 2014; 17:519-27. [PMID: 24673738 DOI: 10.1111/1756-185x.12333] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIM The present study aimed at investigating the effects of Lactobacillus casei 01 supplementation on symptoms and inflammatory biomarkers of rheumatoid arthritis (RA) in women. METHOD In this randomized double-blind clinical trial, female patients with established RA for more than 1 year, 20-80 years of age and body mass index (BMI) lower than 40, who followed stable medication for 3 months prior to the supplementation, were randomly allocated to receive either one capsule containing 10(8) colony forming units (CFU) of L. casei 01, or a placebo for 8 weeks; allocation was stratified by BMI and menopausal status. Disease activity score-28 (DAS28) was calculated, European League Against Rheumatism (EULAR) response was evaluated and the cytokines, interleukin (IL)-1β, IL-6, IL-10, IL-12 and tumor necrosis factor (TNF)-α were measured. RESULTS Thirty patients were recruited in each group; 22 and 24 patients were analyzed in the probiotic and placebo groups, respectively. L. casei 01 supplementation decreased serum high-sensitivity C-reactive protein (hs-CRP) levels, tender and swollen joint counts, global health (GH) score and DAS28 (P < 0.05). More patients in the L. casei 01 group had moderate response to the treatment, based on the EULAR criteria, at the end of the study (P < 0.01). At the end of the study, a significant difference was observed between the two groups for IL-10, IL-12 and TNF-α changes through the study course (P < 0.05), in favor of the probiotic group. No adverse effects were reported for the intervention. CONCLUSION Probiotic supplementation may be an appropriate adjunct therapy for RA patients and help alleviate symptoms and improve inflammatory cytokines.
Collapse
Affiliation(s)
- Beitullah Alipour
- Department of Community Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Vaghef-Mehrabany E, Alipour B, Homayouni-Rad A, Sharif SK, Asghari-Jafarabadi M, Zavvari S. Probiotic supplementation improves inflammatory status in patients with rheumatoid arthritis. Nutrition 2013; 30:430-5. [PMID: 24355439 DOI: 10.1016/j.nut.2013.09.007] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/05/2013] [Accepted: 09/17/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an inflammatory autoimmune disease in which the gut microbiota is altered. Probiotics are microorganisms that can normalize gut microbiota; thus, they may help to alleviate RA symptoms. The objective of the present clinical trial was to assess the effects of probiotic supplementation on disease activity and inflammatory cytokines in patients with RA. METHODS Forty-six patients with RA were assigned into two groups in this randomized, double-blind, placebo-controlled clinical trial. The patients in the probiotic group received a daily capsule that contained a minimum of 10(8) colony-forming units of Lactobacillus casei 01 for 8 wk. The placebo group took capsules filled with maltodextrin for the same time period. Questionnaires, anthropometric measurements, and fasting blood samples were collected, and the participants were assessed by a rheumatologist at baseline and at the end of the trial. RESULTS Disease activity score was significantly decreased by the intervention, and there was a significant difference between the two groups at the end of the study (P < 0.01). Three of the assessed serum proinflammatory cytokines (tumor necrosis factor-α, interleukin-6, and interleukin-12) significantly decreased in the probiotic group (P < 0.05); however, serum levels of interleukin-1 β were not significantly affected by the probiotic (P = 0.22). The serum level of regulatory cytokine (interleukin-10) was increased by the supplementation (P < 0.05). The proportion of interleukin-10 to interleukin-12 was significantly increased in the probiotic group as well. CONCLUSIONS L. casei 01 supplementation improved the disease activity and inflammatory status of patients with RA. Further studies are warranted to confirm these results, and such confirmation may lead to the introduction of probiotics as adjunctive therapy for this population.
Collapse
Affiliation(s)
- Elnaz Vaghef-Mehrabany
- Department of Nutrition, Biochemistry and Diet Therapy, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Beitullah Alipour
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Homayouni-Rad
- Department of Food Science and Technology, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sakineh-Khatoon Sharif
- Rheumatology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Asghari-Jafarabadi
- Road Traffic Injury Prevention Research Center, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sema Zavvari
- Department of Public Health and Management, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Malin M, Verronen P, Korhonen H, Syväoja EL, Salminen S, Mykkänen H, Arvilommi H, Eerola E, Isolauri E. Dietary therapy with Lactobacillus GG, bovine colostrum or bovine immune colostrum in patients with juvenile chronic arthritis: evaluation of effect on gut defence mechanisms. Inflammopharmacology 2010; 5:219-36. [PMID: 17638132 DOI: 10.1007/s10787-997-0001-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/1997] [Accepted: 05/14/1997] [Indexed: 11/28/2022]
Abstract
The effect of dietary therapy with a human Lactobacillus strain GG (ATCC 53103), bovine colostrum, or bovine immune colostrum with specific antibodies against anaerobic intestinal bacteria on gut defence mechanisms were studied in juvenile chronic arthritis. Thirty patients with juvenile chronic arthritis were randomly allocated to receive a freeze-dried powder of Lactobacillus GG, or bovine colostrum, or bovine immune colostrum, for a two-week period. Immunologic and non-immunologic gut defence mechanisms were indirectly investigated in blood and faecal samples. In patients receiving Lactobacillus GG, the median (interquartile range) frequency of immunoglobulin-secreting cells, determined by enzyme-linked immunospot assay, increased in the IgA class from 1840 (690-2530) to 3480 (1030-13 170)/10(6) cells; p=0.02. Likewise the median (interquartile range) frequency of specific antibody-secreting cells against dietary antigens increased during the Lactobacillus GG therapy in the IgM class from 3.8 (1.4-5.0) to 11.2 (5.0-30.0)/10(6) cells; p=0.02. In addition, Lactobacillus GG therapy decreased the median (interquartile range) activity of faecal urease, which has been associated with mucosal tissue damage, from 40.3 (21.7-54.3) to 28.6 (24.5-49.4) nmol. min(-1) (mg protein)(-1); p=0.10, while, in patients receiving bovine colostrum, faecal urease activity increased (from 42.2 to 80.6; p=0.04). All findings were transient. We suggest that gut defence mechanisms are disturbed in juvenile chronic arthritis and we further suggest that orally administered Lactobacillus GG has a potential to reinforce the mucosal barrier mechanisms in juvenile chronic arthritis.
Collapse
Affiliation(s)
- M Malin
- Medical School, University of Tampere and Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Saxelin M. LactobacillusGG—a human probiotic strain with thorough clinical documentation. FOOD REVIEWS INTERNATIONAL 2009. [DOI: 10.1080/87559129709541107] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
19
|
Amital H, Gilburd B, Shoenfeld Y. Probiotic supplementation with Lactobacillus casei (Actimel) induces a Th1 response in an animal model of antiphospholipid syndrome. Ann N Y Acad Sci 2007; 1110:661-9. [PMID: 17911481 DOI: 10.1196/annals.1423.069] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Probiotic fermented milk products have the capacity to modulate many immunological mechanisms. Several attempts have been made to alter the progression of various atopic and inflammatory disorders in which the immune system plays a major role. We studied this issue in an animal model of the antiphospholipid syndrome (APS) by supplementing the animals' daily intake with a probiotic mixture. We studied the effects of nutritional supplementation of a commercial product that consists of 10(8)/ml Lactobacillus casei (Actimel) on Balb/c mice that were immunized with beta-2- glycoprotein (beta2GPI) in order to induce a familiar murine model of APS. As controls, we used similar animals that were fed with either yogurt or sham solution as a supplement. We analyzed the effect of Actimel on the concentrations of interleukin (IL)-10 interferon gamma (IFNgamma) as well as the extent of the primary T cell response to beta2GPI, and the levels of autoantibodies to beta2GPI determined by ELISA. Two weeks after priming (in the hind footpad) of Balb/c mice with beta2GPI, we analyzed the cytokine profile of the animals by measuring the concentration of IL-10 and IFNgamma in the supernatants of lymphocytes that were extracted from the popliteal lymph nodes. Following stimulation with 10 microg/mL of beta2GPI, we noticed significant (P < 0.05) suppression of IL-10 production by the stimulated lymphocytes in the animals fed with Actimel and yogurt in comparison to sham solution (73.42 +/- 29.4, 84.7 +/- 8, 196 +/- 41.62 pg/mL, respectively). Both dairy products enhanced the secretion of IFNgamma from 657 +/- 47.09 pg/mL to 896 +/- 78.1, and 933 +/- 76.7 (P < 0.01), respectively; similarly they also accelerated by a mild degree the level of the T cell primary response to beta2GPI measured by [3H]thymidine incorporation. The level of autoantibodies to beta2GPI was suppressed in mice fed with actimel and yogurt in a significant manner (P < 0.05). Actimel as well as yogurt confer an immunological impact on Balb/c mice immunized with beta2GPI. Actimel was able not only to enhance IFNgamma secretion but also to inhibit IL-10 production.
Collapse
Affiliation(s)
- Howard Amital
- Department of Medicine D, Meir Medical Center, Tshernichovsky 59, Kfar-Saba, 44281, Israel.
| | | | | |
Collapse
|
20
|
Gueimonde M, Ouwehand A, Huhtinen H, Salminen E, Salminen S. Qualitative and quantitative analyses of the bifidobacterial microbiota in the colonic mucosa of patients with colorectal cancer, diverticulitis and inflammatory bowel disease. World J Gastroenterol 2007; 13:3985-9. [PMID: 17663515 PMCID: PMC4171173 DOI: 10.3748/wjg.v13.i29.3985] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To characterize the bifidobacterial microbiota of the colonic mucosa in patients with colon cancer, inflammatory bowel disease or diverticulitis.
METHODS: A sample of the distal colonic mucosa was taken during surgery from a total of 34 patients, twenty-one with diagnosed colorectal cancer, nine with diverticulitis and four with inflammatory bowel disease, requiring surgery for their condition. Bacterial DNA was extracted from the resected mucosal samples and bifidobacterial mucosa-associated microbiota was qualitatively and quantitatively determined by means of qualitative and quantitative PCR.
RESULTS: Bifidobacteria were found in 100% of the samples from patients with diverticulitis or IBD and a 76% of those suffering colon cancer. The species B. longum and B. bifidum were the most widely found, followed by B. animalis, B. catenulatum and B. adolescentis. B. breve, B. dentium and B. angulatum were not detected in any sample. A significantly higher occurrence of B. longum was observed in patients with diverticulitis than in those with colon cancer or IBD (100%, 62% and 75%, respectively, P < 0.05). Similar results were obtained for B. animalis (56%, 0% and 25%, P < 0.05), while B. adolescentis was only found in the mucosa from patients with colon cancer (5 out of 21, 24%). At the quantitative level, patients with colon cancer or IBD showed lower counts of total Bifidobacterium (4.94 and 5.91 vs 6.96 log Cells/sample, respectively, P < 0.05) and of the species B. longum (4.05 and 4.79 vs 6.76, P < 0.05) than those with diverticulitis.
CONCLUSION: Aberrancies in mucosa associated microbiota are present in different intestinal diseases. This may indicate a role of the microbiota in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Miguel Gueimonde
- Instituto de Productos Lacteos de Asturias (IPLA-CSIC). Ctra. Infiesto s/n, 33300, Villaviciosa, Asturias, Spain.
| | | | | | | | | |
Collapse
|
21
|
Farnworth ER, Chouinard YP, Jacques H, Venkatramanan S, Maf AA, Defnoun S, Jones PJH. The effect of drinking milk containing conjugated linoleic acid on fecal microbiological profile, enzymatic activity, and fecal characteristics in humans. Nutr J 2007; 6:15. [PMID: 17620127 PMCID: PMC1949403 DOI: 10.1186/1475-2891-6-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 07/09/2007] [Indexed: 11/16/2022] Open
Abstract
Background The primary objective was to determine whether consumption of conjugated linoleic acids (CLAs) affected the fecal microbiota composition, fecal enzyme activity or fecal composition. Methods Human subjects consumed (1 L/day) cows' milk (4% fat) containing (5 mg/g fat) cis-9, trans-11 CLA (CONT), (32 mg/g fat) cis-9, trans-11 CLA (NAT) and (32 mg/g fat) trans-10, cis-12 CLA and cis-9, trans-11 CLA (SYN) for 8 weeks, in addition to their normal diet. Milk feeding periods were separated by 4 week washout periods. Fecal samples were obtained at the beginning (day 0) and the end (day 56) of each milk feeding period. Fecal samples were analysed for microbiological profile, enzyme activity, pH and short chain fatty acid content. Results Samples taken at day 0 and day 56 indicated that the numbers of lactobacilli and bifidobacteria significantly decreased after consumption of all experimental milks; total aerobes, total anaerobes, enterobacteria, and enterococci + streptococci did not change. At day 56, the activities of β-glucosidase, nitroreductase, and urease enzymes had decreased compared to samples taken on day 0 for all treatments. β-glucuronidase activity did not change. Fecal pH and ammonia content did not change. Conclusion It was concluded that observed changes could have been attributed to increased milk intake; no differences could be attributed to consumption of the different CLAs.
Collapse
Affiliation(s)
- Edward R Farnworth
- Food Research and Development Centre, Agriculture Canada, Saint Hyacinthe, Quebec, Canada
| | - Yvan P Chouinard
- Departments of Animal Science and Food Science and Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Helene Jacques
- Departments of Animal Science and Food Science and Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Sudha Venkatramanan
- School of Dietetics and Human Nutrition, McGill University, Montreal, Quebec, Canada
| | - Akier A Maf
- Food Research and Development Centre, Agriculture Canada, Saint Hyacinthe, Quebec, Canada
| | - Sabrina Defnoun
- Food Research and Development Centre, Agriculture Canada, Saint Hyacinthe, Quebec, Canada
| | - Peter JH Jones
- School of Dietetics and Human Nutrition, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Abstract
Despite the swift escalation in research regarding the use of live bacterial cells for therapeutic purposes, the prophylactic and curative use of probiotic microorganisms still remains a wide and controversial field. In addition, the acknowledgement that live bacterial cells can be genetically engineered to synthesise products that have therapeutic potential has generated substantial interest among clinicians and health professionals. Clinical trials have increasingly provided an insightful scientific derivation for the use of live bacterial cells in medicinal practice in diseases such as diarrhoea, cancer, Crohn's disease, enhancement of the host's immune response, and numerous other diseases. A key constraint in the use of live bacterial cells, however, is the complexity of delivering them to the correct target sites. Oral delivery of free live cells, lyophilised cells and immobilised cells has been attempted, but with restricted success, chiefly because bacterial cells are unable to survive passage through the gastrointestinal tract in sufficient dosage. On many occasions, when given orally, these cells have been found to provoke immunogenic responses that are undesirable. Recent studies show that these problems can be overcome by delivering live bacterial cells using artificial cell microcapsules. This review abridges recent developments in the therapeutic use of live bacterial cells, addresses the potential and restrictions for their application in therapy, and provides insights into the future course of this emerging therapy.
Collapse
Affiliation(s)
- Satya Prakash
- Department of Biomedical Engineering and Artificial Cells, Organs Research Centre, Faculty of Medicine, McGill University, Montreal, Québec, H3A 2B4, Canada.
| | | |
Collapse
|
23
|
Baharav E, Mor F, Halpern M, Weinberger A. Lactobacillus GG bacteria ameliorate arthritis in Lewis rats. J Nutr 2004; 134:1964-9. [PMID: 15284384 DOI: 10.1093/jn/134.8.1964] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Probiotic bacteria have beneficial effects in infectious and inflammatory diseases, principally in bowel disorders. In the case of chronic progressive autoimmune arthritides, a major goal of treatment is to reduce inflammation. We hypothesized that probiotic bacteria would ameliorate inflammation found in arthritis models. To assess this effect, Lewis rats were injected with 50 microg bovine alpha-tropomyosin (TRM) or complete Freund's adjuvant (CFA) to induce tropomyosin arthritis (TA) or adjuvant arthritis (AA), respectively. In both models, the rats were divided into 6 groups and fed 0.5 mL/d of the following suspensions: 1) heat-killed Lactobacillus GG (LGG) bacteria; 2) live LGG, both 10(11) colony-forming units (cfu)/L; 3) sterilized milk; 4) plain yogurt; 5) yogurt containing 10(11) cfu/L LGG; or 6) sterilized water. In the disease-prevention experiments, feeding started 1 wk before or after disease induction. In the therapeutic experiments, feeding was initiated at the onset of clinical arthritis. In all experiments, there were significant interactions between time and treatment (P < 0.001), except for milk, which had no effect in the therapeutic experiment. Histologically, rats fed yogurt containing LGG had a milder inflammation in all experiments (P < 0.05), whereas rats fed plain yogurt exhibited a moderate inflammatory score only in the prevention experiments. Anti-TRM antibody titers were not affected by any of the treatments in any of the experiments. Ingestion of live or heat-killed human LGG had a clinically beneficial effect on experimental arthritis. Our observation of the remarkable preventive and curative effect on arthritis using commercial yogurts containing lactobacilli, especially LGG, suggests the need for investigation of these agents in arthritic patients.
Collapse
Affiliation(s)
- Ehud Baharav
- Department of Medicine B, Rabin Medical Center, and Laboratory of Physiopathology of Joints and Inflammation, Felsenstein Medical Research Center, Petah Tiqva, Israel
| | | | | | | |
Collapse
|
24
|
Mechanick JI, Brett EM, Chausmer AB, Dickey RA, Wallach S. American Association of Clinical Endocrinologists Medical Guidelines for the Clinical Use of Dietary Supplements and Nutraceuticals. Endocr Pract 2003; 9:417-70. [PMID: 14583426 DOI: 10.4158/ep.9.5.417] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
25
|
Abstract
Several clinical studies have demonstrated the therapeutic and/or prophylactic efficacy of specific probiotics against acute viral gastroenteritis and antibiotic-associated diarrhoea (including Clostridium difficile infection). Emerging evidence also suggests beneficial effects against Helicobacter pylori infection. The evidence of efficacy against traveller's diarrhoea remains, however, inconclusive. The precise mechanisms by which probiotics potentiate host gastrointestinal defences and mediate protection are not fully known. There is evidence to suggest, however, that probiotics might contribute to host defence by reinforcing non-immunological defences and stimulating both specific and non-specific host immune responses. Little is known about the relative importance of the probiotic-stimulated mechanisms in host protection. This review summarises the evidence for the anti-infective effects of probiotics and discusses the effect of orally delivered probiotics on non-immunological and immunological defence mechanisms in the host, especially in the gastrointestinal tract.
Collapse
Affiliation(s)
- Harsharnjit S Gill
- Institute of Food, Nutrition and Human Health, Massey University, Private Bag 11222, Palmerston North, New Zealand.
| |
Collapse
|
26
|
Abstract
Evidence for the role of probiotics in maintenance of health or prevention of disease is mounting and is supported in some cases by blinded, placebo-controlled human trials. Today, in an era of antibiotic-resistant pathogens and other looming microbial threats, the value of prevention of infection is recognized. Probiotics may play an important role in helping the body protect itself from infection, especially along the colonized mucosal surfaces of the gastrointestinal tract. Probiotic products are available in many different forms worldwide, including pills, powders, foods, and infant formula. In some cases, general health claims are made that cannot be substantiated for the specific strains and levels being used and consumers must therefore beware.
Collapse
Affiliation(s)
- Mary Ellen Sanders
- Dairy and Food Culture Technologies, 7119 S. Glencoe Ct., Centennial, CO 80122-2526, USA
| |
Collapse
|
27
|
Brigidi P, Vitali B, Swennen E, Bazzocchi G, Matteuzzi D. Effects of probiotic administration upon the composition and enzymatic activity of human fecal microbiota in patients with irritable bowel syndrome or functional diarrhea. Res Microbiol 2001; 152:735-41. [PMID: 11686387 DOI: 10.1016/s0923-2508(01)01254-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In a clinical trial, 10 patients suffering from irritable bowel syndrome or functional diarrhea were administered the probiotic preparation VSL-3. Preliminary results indicated that administration of VSL-3 improved the clinical picture and changed the composition and biochemistry of fecal microbiota. Titer variations of intestinal bacterial groups were evaluated by culture and PCR techniques. A significant increase in lactobacilli, bifidobacteria and Streptococcus thermophilus was observed as a consequence of probiotic treatment, while enterococci, coliforms, Bacteroides and Clostridium perfringens did not change significantly. The strains Bifidobacterium infantis Y1 and Bifidobacterium breve Y8, included in VSL-3, were specifically detected in feces of patients treated with the probiotic by using strain-specific PCR primers. In addition, fecal beta-galactosidase increased and urease activities decreased as a result of changes in the intestinal microbiota induced by VSL-3 administration.
Collapse
Affiliation(s)
- P Brigidi
- Dipartimento di Scienze Farmaceutiche, Università di Bologna, Italy.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Western civilization is facing a progressive increase in immune-mediated, gut-related health problems, such as allergies and autoimmune and inflammatory diseases, and genetic factors are an unlikely explanation for these rapid increases in disease incidence. Two environmental factors that relate to the modern lifestyle in Western societies are hygiene and nutrition. There has been a decline in the incidence of microbial stimulation by infectious diseases as a result of improved hygiene, vaccination, and antimicrobial medication. In the past, methods of food preservation involved either the natural fermentation or drying of foods; thus, the human diet once contained several thousand times more bacteria than it does today. The development of probiotic, functional foods aims to "kill two birds with one stone," which is accomplished by providing a microbial stimulus to the host immune system by means of beneficial live microorganism cultures that are characteristic of the healthy, human gut microflora, ie, probiotics. Probiotic bacteria were shown to reinforce the different lines of gut defense, which are immune exclusion, immune elimination, and immune regulation. They were also shown to stimulate nonspecific host resistance to microbial pathogens, thereby aiding in pathogen eradication. Consequently, the best documented clinical application of probiotics is in the treatment of acute diarrhea. In humans, documented effects were reported for the alleviation of intestinal inflammation, normalization of gut mucosal dysfunction, and down-regulation of hypersensitivity reactions. These data show that probiotics promote endogenous host defense mechanisms. Thus, modification of gut microflora by probiotic therapy may offer a therapeutic potential in clinical conditions associated with gut-barrier dysfunction and inflammatory response.
Collapse
Affiliation(s)
- E Isolauri
- Department of Pediatrics, University of Turku, Finland.
| |
Collapse
|
29
|
Abstract
Clinical studies have shown that certain probiotics may be useful in treating a variety of diarrheal disorders, including rotavirus diarrhea, antibiotic-associated diarrhea, Clostridium difficile diarrhea, and traveler's diarrhea. New data suggest that probiotics might be useful in controlling inflammatory diseases, treating and preventing allergic diseases, preventing cancer, and stimulating the immune system, which may reduce the incidence of respiratory disease. Different modes of administering probiotics are currently being investigated, which may ultimately lead to the widespread use of probiotics in functional foods. It is important that such practices be directed by carefully controlled clinical studies published in peer-reviewed journals.
Collapse
Affiliation(s)
- J A Vanderhoof
- Department of Pediatric Gastroenterology and Nutrition, University of Nebraska/Creighton University, Omaha, USA.
| |
Collapse
|
30
|
Isolauri E, Sütas Y, Kankaanpää P, Arvilommi H, Salminen S. Probiotics: effects on immunity. Am J Clin Nutr 2001; 73:444S-450S. [PMID: 11157355 DOI: 10.1093/ajcn/73.2.444s] [Citation(s) in RCA: 514] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The gastrointestinal tract functions as a barrier against antigens from microorganisms and food. The generation of immunophysiologic regulation in the gut depends on the establishment of indigenous microflora. This has led to the introduction of novel therapeutic interventions based on the consumption of cultures of beneficial live microorganisms that act as probiotics. Among the possible mechanisms of probiotic therapy is promotion of a nonimmunologic gut defense barrier, which includes the normalization of increased intestinal permeability and altered gut microecology. Another possible mechanism of probiotic therapy is improvement of the intestine's immunologic barrier, particularly through intestinal immunoglobulin A responses and alleviation of intestinal inflammatory responses, which produce a gut-stabilizing effect. Many probiotic effects are mediated through immune regulation, particularly through balance control of proinflammatory and anti-inflammatory cytokines. These data show that probiotics can be used as innovative tools to alleviate intestinal inflammation, normalize gut mucosal dysfunction, and down-regulate hypersensitivity reactions. More recent data show that differences exist in the immunomodulatory effects of candidate probiotic bacteria. Moreover, distinct regulatory effects have been detected in healthy subjects and in patients with inflammatory diseases. These results suggest that specific immunomodulatory properties of probiotic bacteria should be characterized when developing clinical applications for extended target populations.
Collapse
Affiliation(s)
- E Isolauri
- Department of Pediatrics, the University of Turku, Turku, Finland.
| | | | | | | | | |
Collapse
|
31
|
Vanderhoof JA, Young RJ. The role of probiotics in the treatment of intestinal infections and inflammation. Curr Opin Gastroenterol 2001; 17:58-62. [PMID: 17031151 DOI: 10.1097/00001574-200101000-00011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Clinical studies have established that certain probiotics are useful in a variety of intestinal disorders, including viral diarrhea, antibiotic-associated diarrhea, Clostridium difficile-associated diarrhea, and traveler's diarrhea. Evolving data suggest that probiotics might be useful in the control of inflammatory diseases, treatment and prevention of allergic diseases, cancer prevention, and stimulation of the immune system, which may result in the reduction in respiratory disease. A review of current data regarding the most common probiotics is presented. It is important that probiotic use be directed by well-designed, placebo-controlled clinical studies published in peer-reviewed journals.
Collapse
Affiliation(s)
- J A Vanderhoof
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.
| | | |
Collapse
|
32
|
Abstract
Oral consumption of probiotic bacteria has the potential to support the health of American consumers. This paper will discuss the rationale of the probiotic theory, several health targets for probiotic bacteria, probiotic products in the U.S. and, finally, issues pertaining to communication about probiotic products to the consumer.
Collapse
Affiliation(s)
- M E Sanders
- Dairy and Food Culture Technologies, Littleton, CO 80122-2526, USA
| |
Collapse
|
33
|
Grönlund MM, Salminen S, Mykkänen H, Kero P, Lehtonen OP. Development of intestinal bacterial enzymes in infants--relationship to mode of delivery and type of feeding. APMIS 1999; 107:655-60. [PMID: 10440061 DOI: 10.1111/j.1699-0463.1999.tb01455.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To evaluate the development of intestinal flora in young infants, and especially to estimate the influence of mode of delivery and type of feeding on the establishment of intestinal microflora, faecal flora was studied indirectly by measuring prospectively the faecal bacterial enzyme activities (beta-glucosidase, beta-glucuronidase and urease) in 29 full-term, healthy infants during the first 6 months of life. Mode of delivery had no influence on the faecal enzyme activities. In contrast, infants receiving formula feeds were more often urease positive at 1-2 months of age (70% vs 25%, p=0.043) and had higher median activity of beta-glucuronidase at 6 months of age (0.90 and 0.19 nmoles/mg protein x min, p= 0.0043) than exclusively breast-fed infants. Through indirect methods to measure the development of a faecal microflora our results indicate that the type of milk that infants receive during the first months of life may have an important role in the development of intestinal flora.
Collapse
Affiliation(s)
- M M Grönlund
- Department of Pediatrics, Turku University Central Hospital, Finland
| | | | | | | | | |
Collapse
|
34
|
Pessi T, Sütas Y, Marttinen A, Isolauri E. Probiotics reinforce mucosal degradation of antigens in rats: implications for therapeutic use of probiotics. J Nutr 1998; 128:2313-8. [PMID: 9868175 DOI: 10.1093/jn/128.12.2313] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The effects of probiotics, administered with different diets, i.e., unhydrolyzed or hydrolyzed dietary antigens, on macromolecular degradation in the gut mucosa were studied. Rat pups were divided into five feeding groups at the age of 14 d. In addition to maternal milk, the milk group was gavaged daily with cows' milk and the hydrolysate group with extensively hydrolyzed whey formula, while controls received sterile saline. In addition to these diets, the milk-GG group and the hydrolysate-GG group were given probiotic bacteria, Lactobacillus GG ATCC 53103 (10(10) colony-forming units per day). At 21 d, the absorption of macromolecules, horseradish peroxidase and beta-lactoglobulin across patch-free jejunal segments was studied in Ussing chambers. The degree of macromolecular degradation was studied by means of HPLC gel filtration. The absorption rate of intact horseradish peroxidase differed among the feeding groups (P = 0.038). This was due to the high median (interquartile range) absorption of intact horseradish peroxidase (ng x h-1 x cm-2) in the milk group [255 (14-1332)] and supplementation with L. GG in the milk-GG group [35 (8-233)] restoring the status to the control level [22 (0-116)]. A parallel effect was seen in the hydrolysate group [100 (9-236)] vs. the hydrolysate-GG group [1 (0- 13)]. A gel filtration study confirmed that larger molecules were absorbed across the mucosa in the milk group compared to the other groups. The absorption of degraded horseradish peroxidase differed between the feeding groups (P = 0. 005). L. GG had a distinct effect when administered with unhydrolyzed, native protein vs. hydrolyzed protein: it increased absorption of degraded horseradish peroxidase in the milk-GG group [7310 (4763-8228)] vs. the milk group [3726 (2423-5915)], while reducing it in the hydrolysate-GG group [2051 (1463-2815)] vs. the hydrolysate group [4573 (3759-9620)]. Our results showed that probiotics not only restore aberrant macromolecular transport, but they also have a specific effect on mucosal degradation depending on dietary antigen: adjuvant-like properties (unhydrolyzed antigen) and immunosuppressive-like properties (hydrolyzed antigen). The antigenicity of the diet therefore should be taken into consideration, when introducing novel probiotic functional foods for the management of gastrointestinal disorders.
Collapse
Affiliation(s)
- T Pessi
- Department of Pediatrics, University of Turku, Finland
| | | | | | | |
Collapse
|
35
|
Abstract
Probiotics appear to be useful in the prevention or treatment of several gastrointestinal disorders, including infectious diarrhea, antibiotic diarrhea, and traveler's diarrhea. Results of preliminary human and animal studies suggest that patients with inflammatory diseases, and even irritable bowel syndrome, may benefit from probiotic therapy. Probiotics represent an exciting therapeutic advance, although much investigation must be undertaken before their role in gastroenterology is clearly delineated. Questions related to probiotic origin, survivability, and adherence are all important considerations for further study. More important, each probiotic proposed must be studied individually and extensively to determine its efficacy and safety in each disorder for which its use may be considered.
Collapse
Affiliation(s)
- J A Vanderhoof
- Department of Pediatrics, University of Nebraska, Omaha, USA
| | | |
Collapse
|
36
|
Salminen S, Isolauri E, Salminen E. Clinical uses of probiotics for stabilizing the gut mucosal barrier: successful strains and future challenges. Antonie Van Leeuwenhoek 1996; 70:347-58. [PMID: 8992950 DOI: 10.1007/bf00395941] [Citation(s) in RCA: 223] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Probiotic bacteria are used to treat disturbed intestinal microflora and increased gut permeability which are characteristic to many intestinal disorders. Examples include children with acute rotavirus diarrhoea, subjects with food allergy, subjects with colonic disorders and patients undergoing pelvic radiotherapy and sometimes changes associated with colon cancer development. In all such disease states altered intestinal microflora, impaired gut barrier and different types of intestinal inflammation are present. Successful probiotic bacteria are able to survive gastric conditions and colonize the intestine, at least temporarily, by adhering to the intestinal epithelium. Such probiotic microorganisms appear to be promising candidates for the treatment of clinical conditions with abnormal gut microflora and altered gut mucosal barrier functions. They are also promising ingredients to future functional foods and clinical foods for specific disease states provided that basic requirements for strains and clinical studies are carefully followed.
Collapse
Affiliation(s)
- S Salminen
- Department of Biochemistry and Food Chemistry, University of Turku, Finland
| | | | | |
Collapse
|