51
|
Sommermeyer H, Piątek J. Synbiotics as Treatment for Irritable Bowel Syndrome: A Review. Microorganisms 2024; 12:1493. [PMID: 39065261 PMCID: PMC11278745 DOI: 10.3390/microorganisms12071493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Irritable bowel syndrome is a persistent disturbance of the function of the gastrointestinal tract with a prevalence of about 11.2% in the population at large. While the etiology of the disorder remains unclear, there is mounting evidence that the disturbance of the gut microbiota is at least one contributing factor. This insight resulted in clinical trials investigating the therapeutic effects of products containing probiotic microorganisms. Most studies with IBS patients have evaluated the therapeutic effects of mono- and multi-strain probiotics, but only a few studies have investigated the efficacy of synbiotics (combinations of probiotic bacteria and one or more prebiotic components). This review summarizes the results from eight randomized, placebo-controlled clinical trials that investigated the efficacy of synbiotic preparations (three mono-strain and five multi-strain products) in adult IBS patients. While data remain sparse, some of the surveyed clinical trials have demonstrated interesting efficacy results in IBS patients. To allow a judgment of the role played by synbiotics in the treatment of IBS patients, more high-quality clinical trials are needed.
Collapse
Affiliation(s)
- Henning Sommermeyer
- Department of Health Sciences, Calisia University, Nowy Swiat 4, 62-800 Kalisz, Poland;
| | | |
Collapse
|
52
|
Alqaydi TK, Bedir AS, Abu-Elsaoud AM, El-Tarabily KA, Al Raish SM. An Assessment of the Knowledge, Attitude, and Practice of Probiotics and Prebiotics among the Population of the United Arab Emirates. Foods 2024; 13:2219. [PMID: 39063303 PMCID: PMC11276527 DOI: 10.3390/foods13142219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Probiotics and prebiotics offer a range of advantageous effects on human health. The knowledge, attitudes, and practices (KAP) of individuals can impact their inclination to consume probiotics and prebiotics. The main objective of this study was to examine the KAP of the people in the United Arab Emirates (UAE) about probiotics and prebiotics consumption. Additionally, the study aimed to assess the impact of KAP and sociodemographic factors on the use of probiotics and prebiotics. In order to accomplish this objective, a verified online questionnaire was used with a five-point Likert scale and distributed using an online platform (Google Forms). A cross-sectional research, non-probability sampling was implemented, and G*Power statistical power analysis was used to estimate a sample size of 385 participants. A total of 408 replies were gathered. The population under study consisted of residents in the UAE between the ages of 18 to 64 years old, excluding populations under the age of 18 and those living outside the UAE. A total of 392 participants met the criteria for inclusion in this study. The research ethics committees of UAE University granted the study approval (ERSC_2024_4359), and the validity of the findings was confirmed through face-to-face interviews with around 50 individuals and a Cronbach's alpha test with result of 0.84. The statistical software SPSS version 29.0 for Mac OS was utilized to examine the relationships between KAP variables, including Chi-square tests and Pearson's correlation coefficients. The tests were selected based on their capacity to handle categorical and continuous data, respectively. The female population was 85.2% of the total, while the male population accounts for 14.8%. The age distribution of participants shows that the largest proportion, 68.4%, falls within the 18-24 age range. Out of the participants, 61.5% held a bachelor's degree. Most of the participants, 56.4%, were students, while 29.1% were employees. The average results indicate a significant inclination towards probiotics and prebiotics, as demonstrated by the scores above the midpoint for the six knowledge questions (M = 2.70), six attitude questions (M = 3.10), and six practice questions (M = 3.04). Several studies have examined this phenomenon; however, additional research comparing individuals in the UAE is necessary to fully comprehend the influence of KAP on the consumption of probiotics and prebiotics in the UAE.
Collapse
Affiliation(s)
- Turfa K. Alqaydi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (T.K.A.); (K.A.E.-T.)
| | - Alaa S. Bedir
- Department of Nutrition, College of Medicine and Health Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Abdelghafar M. Abu-Elsaoud
- Department of Botany and Microbiology, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt;
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Khaled A. El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (T.K.A.); (K.A.E.-T.)
| | - Seham M. Al Raish
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (T.K.A.); (K.A.E.-T.)
| |
Collapse
|
53
|
Tabrizi E, Pourteymour Fard Tabrizi F, Mahmoud Khaled G, Sestito MP, Jamie S, Boone BA. Unraveling the gut microbiome's contribution to pancreatic ductal adenocarcinoma: mechanistic insights and therapeutic perspectives. Front Immunol 2024; 15:1434771. [PMID: 39044834 PMCID: PMC11263025 DOI: 10.3389/fimmu.2024.1434771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
The gut microbiome plays a significant role in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), influencing oncogenesis, immune responses, and treatment outcomes. Studies have identified microbial species like Porphyromonas gingivalis and Fusobacterium nucleatum, that promote PDAC progression through various mechanisms. Additionally, the gut microbiome affects immune cell activation and response to immunotherapy, including immune checkpoint inhibitors and CAR-T therapy. Specific microbes and their metabolites play a significant role in the effectiveness of immune checkpoint inhibitors (ICIs). Alterations in the gut microbiome can either enhance or diminish responses to PD-1/PD-L1 and CTLA-4 blockade therapy. Additionally, bacterial metabolites like trimethylamine N-oxide (TMAO) and lipopolysaccharide (LPS) impact antitumor immunity, offering potential targets to augment immunotherapy responses. Modulating the microbiome through fecal microbiota transplantation, probiotics, prebiotics, dietary changes, and antibiotics shows promise in PDAC treatment, although outcomes are highly variable. Dietary modifications, particularly high-fiber diets and specific fat consumption, influence microbiome composition and impact cancer risk. Combining microbiome-based therapies with existing treatments holds potential for improving PDAC therapy outcomes, but further research is needed to optimize their effectiveness.
Collapse
Affiliation(s)
- Eileen Tabrizi
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
- Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Fatemeh Pourteymour Fard Tabrizi
- Department of Community Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran
| | - Gehad Mahmoud Khaled
- Department of Biotechnology, School of Sciences and Engineering, American University in Cairo, New Cairo, Cairo, Egypt
| | - Michael P. Sestito
- Department of Surgery, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Saeid Jamie
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Brian A. Boone
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
- Department of Surgery, West Virginia University School of Medicine, Morgantown, WV, United States
| |
Collapse
|
54
|
Yuan M, Zhang Z, Liu T, Feng H, Liu Y, Chen K. The Role of Nondigestible Oligosaccharides in Alleviating Human Chronic Diseases by Regulating the Gut Microbiota: A Review. Foods 2024; 13:2157. [PMID: 38998662 PMCID: PMC11241040 DOI: 10.3390/foods13132157] [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: 05/25/2024] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024] Open
Abstract
The gut has been a focus of chronic disease research. The gut microbiota produces metabolites that act as signaling molecules and substrates, closely influencing host health. Nondigestible oligosaccharides (NDOs), as a common dietary fiber, play an important role in regulating the structure and function of the gut microbiota. Their mechanism of action is mainly attributed to providing a carbon source as specific probiotics, producing related metabolites, and regulating the gut microbial community. However, due to the selective utilization of oligosaccharides, some factors, such as the type and structure of oligosaccharides, have different impacts on the composition of microbial populations and the production of metabolites in the colon ecosystem. This review systematically describes the key factors influencing the selective utilization of oligosaccharides by microorganisms and elaborates how oligosaccharides affect the host's immune system, inflammation levels, and energy metabolism by regulating microbial diversity and metabolic function, which in turn affects the onset and progress of chronic diseases, especially diabetes, obesity, depression, intestinal inflammatory diseases, and constipation. In this review, we re-examine the interaction mechanisms between the gut microbiota and its associated metabolites and diseases, and we explore new strategies for promoting human health and combating chronic diseases through dietary interventions.
Collapse
Affiliation(s)
- Meiyu Yuan
- State Key Laboratory of Food Science and Resource, Engineering Research Center for Biomass Conversion, Ministry of Education, Nanchang University, Nanchang 330047, China; (M.Y.); (Z.Z.)
| | - Zhongwei Zhang
- State Key Laboratory of Food Science and Resource, Engineering Research Center for Biomass Conversion, Ministry of Education, Nanchang University, Nanchang 330047, China; (M.Y.); (Z.Z.)
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330019, China;
| | - Tongying Liu
- Jiangxi Maternel and Child Health Hospital, Nanchang 330108, China;
| | - Hua Feng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330019, China;
| | - Yuhuan Liu
- State Key Laboratory of Food Science and Resource, Engineering Research Center for Biomass Conversion, Ministry of Education, Nanchang University, Nanchang 330047, China; (M.Y.); (Z.Z.)
- Chongqing Research Institute of Nanchang University, Chongqing 402660, China
| | - Kai Chen
- Shangrao Innovation Institute of Agricultural Technology, College of Life Science, Shangrao Normal University, Shangrao 334001, China
| |
Collapse
|
55
|
Mao Q, Wang X, Cai H, Yang J, Zhang Y, Min W, Qian Q, Zeng Y. Research Progress on the Correlation of Atopic Dermatitis with Gut Microbiota. Clin Cosmet Investig Dermatol 2024; 17:1613-1619. [PMID: 39006130 PMCID: PMC11244069 DOI: 10.2147/ccid.s442551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 06/02/2024] [Indexed: 07/16/2024]
Abstract
Atopic dermatitis (AD) is a common skin disease, the pathogenesis of which has not been fully elucidated. The gut microbiota is the largest micro-ecosystem in the human body that affects the immune system and skin barrier function. Recent studies have shown that in addition to the environmental factors, skin barrier, genetic factors and immune response, gut microbiota disturbance may also cause AD. This review described the correlation of AD with gut microbiota and existing research status of AD treatment via targeting gut microbiota.
Collapse
Affiliation(s)
- Qiuyu Mao
- Department of Dermatology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xinyi Wang
- Department of Dermatology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Haibin Cai
- Department of Dermatology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jingyi Yang
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Yiwen Zhang
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Wei Min
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Qihong Qian
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Yibin Zeng
- Department of Dermatology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
56
|
Chu NHS, Chow E, Chan JCN. The Therapeutic Potential of the Specific Intestinal Microbiome (SIM) Diet on Metabolic Diseases. BIOLOGY 2024; 13:498. [PMID: 39056692 PMCID: PMC11273990 DOI: 10.3390/biology13070498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
Exploring the intricate crosstalk between dietary prebiotics and the specific intestinal microbiome (SIM) is intriguing in explaining the mechanisms of current successful dietary interventions, including the Mediterranean diet and high-fiber diet. This knowledge forms a robust basis for developing a new natural food therapy. The SIM diet can be measured and evaluated to establish a reliable basis for the management of metabolic diseases, such as diabetes, metabolic (dysfunction)-associated fatty liver disease (MAFLD), obesity, and metabolic cardiovascular disease. This review aims to delve into the existing body of research to shed light on the promising developments of possible dietary prebiotics in this field and explore the implications for clinical practice. The exciting part is the crosstalk of diet, microbiota, and gut-organ interactions facilitated by producing short-chain fatty acids, bile acids, and subsequent metabolite production. These metabolic-related microorganisms include Butyricicoccus, Akkermansia, and Phascolarctobacterium. The SIM diet, rather than supplementation, holds the promise of significant health consequences via the prolonged reaction with the gut microbiome. Most importantly, the literature consistently reports no adverse effects, providing a strong foundation for the safety of this dietary therapy.
Collapse
Affiliation(s)
- Natural H. S. Chu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
57
|
Zangirolamo AF, Souza AK, Yokomizo DN, Miguel AKA, da Costa MC, Alfieri AA, Seneda MM. Updates and Current Challenges in Reproductive Microbiome: A Comparative Analysis between Cows and Women. Animals (Basel) 2024; 14:1971. [PMID: 38998083 PMCID: PMC11240322 DOI: 10.3390/ani14131971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The microbiota plays an important role in numerous physiological processes, pathogenesis, development, and metabolism in different animal species. In humans, several studies have demonstrated an association between the vaginal microbiota and fertility rates, and even success in assisted reproduction techniques. In the context of cattle reproduction, although few studies have addressed the microbiota in a healthy state (which is not associated with diseases that affect the reproductive tract of cows), changes in its composition also seem to influence fertility. This review aims to explain the importance of the reproductive microbiota in female bovines and what is available in the literature regarding its possible role in increasing fertility. What are the challenges involved in this process? Future perspectives on its use and manipulation as a selection or intervention tool. Will it be possible to one day extrapolate the findings to reality and apply them in the field? In short, understanding the role of the reproductive microbiota of female bovines can signal the prospect of increasing production, whether of milk or meat, from the same number of animals, as it can optimize reproductive efficiency and perhaps become an allied tool for the economic profitability and sustainability of livestock farming.
Collapse
Affiliation(s)
- Amanda Fonseca Zangirolamo
- National Institute of Science and Technology for Dairy Production Chain (INCT–LEITE), Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.F.Z.); (A.A.A.)
- Laboratory of Animal Reproduction, Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.K.S.); (D.N.Y.); (A.K.A.M.)
| | - Anne Kemmer Souza
- Laboratory of Animal Reproduction, Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.K.S.); (D.N.Y.); (A.K.A.M.)
| | - Deborah Nakayama Yokomizo
- Laboratory of Animal Reproduction, Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.K.S.); (D.N.Y.); (A.K.A.M.)
| | - Ana Karolyne Alves Miguel
- Laboratory of Animal Reproduction, Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.K.S.); (D.N.Y.); (A.K.A.M.)
| | | | - Amauri Alcindo Alfieri
- National Institute of Science and Technology for Dairy Production Chain (INCT–LEITE), Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.F.Z.); (A.A.A.)
| | - Marcelo Marcondes Seneda
- National Institute of Science and Technology for Dairy Production Chain (INCT–LEITE), Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.F.Z.); (A.A.A.)
- Laboratory of Animal Reproduction, Universidade Estadual de Londrina, Londrina 86057-970, PR, Brazil; (A.K.S.); (D.N.Y.); (A.K.A.M.)
| |
Collapse
|
58
|
Popov IV, Koopmans B, Venema K. Modulation of human gut microbiota by linear and branched fructooligosaccharides in an in vitro colon model (TIM-2). J Appl Microbiol 2024; 135:lxae170. [PMID: 38986506 DOI: 10.1093/jambio/lxae170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024]
Abstract
AIMS This study aimed to compare the effects of linear and branched fructooligosaccharides (FOS) extracted from chicory and grass (Lolium perenne), respectively on human microbiota composition, diversity, and metabolism. METHODS AND RESULTS To test the effects of linear and branched FOS on human microbiota we used the artificial in vitro human colon model (TIM-2). Microbiota composition and diversity were assessed by V3-V4 16S rRNA metagenomic sequencing, followed by differential taxa abundance and alpha/beta diversity analyses. SCFA/BCFA production was evaluated by gas chromatography-mass spectrometry. As a result, branched FOS had the most beneficial effects on microbial diversity and metabolite production. Also, branched FOS significantly increased the abundance of commensal bacteria associated with maintaining healthy gut functions and controlling inflammation, such as Butyricicoccus, Erysipelotrichaceae, Phascolarctobacterium, and Sutterella. Linear FOS also significantly increased the abundance of some other commensal gut bacteria (Anaerobutyricum, Lachnospiraceae, Faecalibacterium), but there were no differences in diversity metrics compared to the control. CONCLUSIONS The study revealed that branched FOS had the most beneficial effects compared to the linear FOS in vitro, concerning microbiota modulation, and metabolite production, making this a good candidate for further studies in food biotechnology.
Collapse
Affiliation(s)
- Igor V Popov
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, 5928 SZ Venlo, The Netherlands
- Faculty of Bioengineering and Veterinary Medicine, Don State Technical University, 344000 Rostov-on-Don, Russia
| | | | - Koen Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, 5928 SZ Venlo, The Netherlands
| |
Collapse
|
59
|
Gopalarathinam R, Sankar R, Zhao SS. Role of Anti-Inflammatory Diet and Fecal Microbiota Transplant in Psoriatic Arthritis. Clin Ther 2024; 46:588-596. [PMID: 38862291 DOI: 10.1016/j.clinthera.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE Psoriatic arthritis (PsA) is a chronic inflammatory condition with complex and heterogenous manifestations. Although a myriad of treatment options including biologic medications are available to alleviate symptoms and slow disease progression, there is currently no cure for this condition. There has been a recent emergence of understanding about the relationship between the gut microbiome and immune-mediated inflammatory diseases. This has generated interest in the potential role of dietary interventions, particularly anti-inflammatory diets, and fecal microbiota transplant (FMT) as novel therapeutic approaches. The purpose of this narrative review is to examine the role of an anti-inflammatory diet and FMT in turn and whether their combination may offer alternate approaches for the management of PsA. METHODS Our non-systematic narrative review was informed by a literature search using PubMed and Google Scholar using the terms anti-inflammatory diet, FMT, nutrition supplements, and PsA. Preclinical studies and non-English language articles were excluded when synthesizing the narrative review. FINDINGS Current randomized controlled trials (RCTs) and observational evidence suggest that a hypocaloric diet or Mediterranean diet can help achieve weight loss among PsA patients who are overweight or obese, which in turn reduces inflammation and improves disease activity. However, there is no strong data to support the beneficial effects of intermittent fasting, vitamin supplements, turmeric supplements, probiotics, or omega-3 fatty acid supplements in PsA. Current evidence on the use of FMT in PsA is limited as only one small RCT has been conducted which did not demonstrate efficacy for improving clinical symptoms. IMPLICATIONS Clinicians can consider recommending hypocaloric or Mediterranean diets as an adjunct to standard management of PsA, possibly under the guidance of a dietician. Further research is needed to explore the beneficial effects of the synergistic role of combining an anti-inflammatory diet with FMT in PsA.
Collapse
Affiliation(s)
- Rajesh Gopalarathinam
- Division of Rheumatology, Wrightington Wigan and Leigh Teaching Hospitals NHS Foundation Trust, Wigan, UK.
| | - Reethika Sankar
- Meenakshi Medical College and Research Institute, Meenakshi Nagar, Tamil Nadu, India
| | - Sizheng Steven Zhao
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Science, School of Biological Sciences, Faculty of Biological Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
60
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
61
|
Barrenechea PM. Interaction of the Gut Microbiome With Cancer Treatment. J Adv Pract Oncol 2024; 15:311-319. [PMID: 39328379 PMCID: PMC11424160 DOI: 10.6004/jadpro.2024.15.5.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
The gut microbiome is known to influence health and well-being beyond the gastrointestinal system, including metabolism, mood, and cognitive function. Research on the influence of the gut microbiome on cancer and cancer treatment has expanded in recent decades. This review discusses the effects of the gut microbiome on the pathogenesis of certain cancers, as well as the current guidelines and recommendations for health-care professionals for modifying the gut microbiome in cancer patients currently receiving chemotherapy or immunotherapy. The focus of this review is on five major areas of gut microbiome research (colorectal cancer, melanoma, renal cell carcinoma and non-small cell lung cancer, lymphoma, and acute leukemia) in which therapies, and particularly checkpoint inhibitors, have considerably improved survival outcomes. The relationship between microbial species and therapies to cure malignancies is largely unclear. This review will delineate the relationships being studied and conclusions to draw from the research in these areas thus far.
Collapse
|
62
|
Jadhav HB, Choudhary P, Annapure U, Ramniwas S, Mugabi R, Ahmad Nayik G. The role of sonication in developing synbiotic Beverages: A review. ULTRASONICS SONOCHEMISTRY 2024; 107:106941. [PMID: 38861817 PMCID: PMC11209632 DOI: 10.1016/j.ultsonch.2024.106941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/25/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Synbiotics are a combination of probiotic cells and prebiotic components and this harmonious association has numerous health benefits. Conventional processing technologies use high temperatures for processing which reduces the viability and the final quality of synbiotic beverages. Sonication is a rapidly growing technology in the food processing sector and can be employed for the formulation of synbiotic beverages with improved functionalities. The cavitation events generated during the sonication result in beneficial effects like increased viability of probiotic cells, enhanced bifidogenic characteristics of prebiotic components, less processing time, and high-quality products. The sonication process does not affect the sensory attributes of synbiotic beverages however, it alters the structure of prebiotics thus increasing the access by the probiotics. These positive effects are solely dependent on the type of ultrasound process and the ultrasound operating parameters. The review aims to provide information on the technological aspects of ultrasound, a brief about synbiotics, details on the ultrasound process used for the formulation of synbiotics, the influence of ultrasound operating parameters, and a focus on the research gap.
Collapse
Affiliation(s)
- Harsh B Jadhav
- Department of Food Engineering and Technology, Institute of Chemical Technology, Matunga, Mumbai 400019, India; PIHM, Unit UMET, INRAE, 369 Rue Jules Guesde 59650, Villeneuve d'Ascq -59650, France.
| | - Pintu Choudhary
- Department of Food Technology, CBL Government Polytechnic, Bhiwani, Haryana, India.
| | - Uday Annapure
- Department of Food Engineering and Technology, Institute of Chemical Technology, Matunga, Mumbai 400019, India
| | - Seema Ramniwas
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, Punjab, India
| | - Robert Mugabi
- Department of Food Technology and Nutrition, Makerere University, Kampala, Uganda.
| | - Gulzar Ahmad Nayik
- Department of Food Science & Technology, Govt. Degree College, Shopian 192303, J&K, India.
| |
Collapse
|
63
|
Chatterjee A, Kumar S, Roy Sarkar S, Halder R, Kumari R, Banerjee S, Sarkar B. Dietary polyphenols represent a phytotherapeutic alternative for gut dysbiosis associated neurodegeneration: A systematic review. J Nutr Biochem 2024; 129:109622. [PMID: 38490348 DOI: 10.1016/j.jnutbio.2024.109622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 03/17/2024]
Abstract
Globally, neurodegeneration and cerebrovascular disease are common and growing causes of morbidity and mortality. Pathophysiology of this group of diseases encompasses various factors from oxidative stress to gut microbial dysbiosis. The study of the etiology and mechanisms of oxidative stress as well as gut dysbiosis-induced neurodegeneration in Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, autism spectrum disorder, and Huntington's disease has recently received a lot of attention. Numerous studies lend credence to the notion that changes in the intestinal microbiota and enteric neuroimmune system have an impact on the initiation and severity of these diseases. The prebiotic role of polyphenols can influence the makeup of the gut microbiota in neurodegenerative disorders by modulating intracellular signalling pathways. Metabolites of polyphenols function directly as neurotransmitters by crossing the blood-brain barrier or indirectly via influencing the cerebrovascular system. This assessment aims to bring forth an interlink between the consumption of polyphenols biotransformed by gut microbiota which in turn modulate the gut microbial diversity and biochemical changes in the brain. This systematic review will further augment research towards the association of dietary polyphenols in the management of gut dysbiosis-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Amrita Chatterjee
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Satish Kumar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Suparna Roy Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Ritabrata Halder
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Rashmi Kumari
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Biswatrish Sarkar
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Ranchi, Jharkhand, India.
| |
Collapse
|
64
|
Yemula N, Sheikh R. Gut microbiota in axial spondyloarthritis : genetics, medications and future treatments. ARP RHEUMATOLOGY 2024; 3:216-225. [PMID: 39243363 DOI: 10.63032/wuii1201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Axial spondyloarthritis, also referred to as ankylosing spondylitis, is a chronic inflammatory condition that predominantly affects the axial spine but may also present with peripheral arthritis. It falls within the umbrella of disorders known as spondyloarthropathies. In addition to axial spondyloarthritis, this group includes psoriatic arthritis, enteropathic arthritis, reactive arthritis, and undifferentiated spondyloarthropathy, with axial spondyloarthritis being one of the most common. The overall mechanisms underlying the development of axial spondyloarthritis are complex and multifactorial. There is a significant and well-recognized association between axial spondyloarthritis and the HLA-B27 gene, but there have also been non-HLA genes identified in the disease process, as well as certain inflammatory cytokines that play a role in the inflammatory process, such as tumor necrosis factor (TNF). More recently, there has been research and new evidence linking changes in the gut microbiota to the disease process of axial spondyloarthritis. Research into the role of the gut microbiota and gut dysbiosis is a large, ever-growing field. It has been associated with a multitude of conditions, including axial spondyloarthritis. This mini-review highlights the symbiotic relationship of the gut microbiota with the pathogenesis, therapeutic agents and future treatments of axial spondyloarthritis.
Collapse
|
65
|
Bock PM, Martins AF, Schaan BD. Understanding how pre- and probiotics affect the gut microbiome and metabolic health. Am J Physiol Endocrinol Metab 2024; 327:E89-E102. [PMID: 38809510 DOI: 10.1152/ajpendo.00054.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
The gut microbiome, a complex assembly of microorganisms, significantly impacts human health by influencing nutrient absorption, the immune system, and disease response. These microorganisms form a dynamic ecosystem that is critical to maintaining overall well-being. Prebiotics and probiotics are pivotal in regulating gut microbiota composition. Prebiotics nourish beneficial bacteria and promote their growth, whereas probiotics help maintain balance within the microbiome. This intricate balance extends to several aspects of health, including maintaining the integrity of the gut barrier, regulating immune responses, and producing metabolites crucial for metabolic health. Dysbiosis, or an imbalance in the gut microbiota, has been linked to metabolic disorders such as type 2 diabetes, obesity, and cardiovascular disease. Impaired gut barrier function, endotoxemia, and low-grade inflammation are associated with toll-like receptors influencing proinflammatory pathways. Short-chain fatty acids derived from microbial fermentation modulate anti-inflammatory and immune system pathways. Prebiotics positively influence gut microbiota, whereas probiotics, especially Lactobacillus and Bifidobacterium strains, may improve metabolic outcomes, such as glycemic control in diabetes. It is important to consider strain-specific effects and study variability when interpreting these findings, highlighting the need for further research to optimize their therapeutic potential. The aim of this report is therefore to review the role of the gut microbiota in metabolic health and disease and the effects of prebiotics and probiotics on the gut microbiome and their therapeutic role, integrating a broad understanding of physiological mechanisms with a clinical perspective.
Collapse
Affiliation(s)
- Patricia M Bock
- Pharmacology, Institute of Basic Science, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Andreza F Martins
- Microbiology, Department of Microbiology, Immunology, and Parasitology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Beatriz D Schaan
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
66
|
Li Y, Jiang Y, Zhang Z, Loake VIP, Bao X, Loake GJ. Improvement of both human and animal memory by synergy between fructooligosaccharide and L-theanine function establishing a safe and effective food supplement. Food Sci Nutr 2024; 12:4966-4980. [PMID: 39055226 PMCID: PMC11266938 DOI: 10.1002/fsn3.4145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/24/2024] [Accepted: 03/19/2024] [Indexed: 07/27/2024] Open
Abstract
Aging is classically associated with a decline of cognitive abilities, especially in relation to memory. While the development of potential treatments for neurodegenerative diseases has been in sharp focus, mild cognitive impairment (MCI), a form of age-related memory loss, in the absence of severe functional impairment, a condition experienced by many healthy adults, has received relatively little attention. Advances in this space would make significant contributions to the goal of healthy aging and may also help promote cognitive performance across the wider population. The individual action of either fructooligosaccharide (FOS) or L-theanine, both natural plant-derived molecules, has been tentatively linked with improvements in cognition, but our understanding remains far from complete. We therefore determined the effect of different dose combinations of FOS and L-theanine (termed MT-01/GBL-Memory1) in mice against FOS and L-theanine monotherapy. FOS and L-theanine were found to synergistically enhance murine memory in our animal tests at a dose of 100 mg/kg (coefficient of drug interaction (CDI) < 1). In a subsequent human trial, we demonstrated that MT-01 improved the memory of healthy adults after 1 month of consumption. Our results suggest that a combination of FOS and L-theanine synergistically enhances murine memory within a specific dose range. We show that this plant natural product regimen also improves human memory in a population of healthy adults. MT-01 therefore represents a novel, safe, and effective dietary supplement to promote human memory and cognition.
Collapse
Affiliation(s)
- Yuan Li
- Green Bioactives Limited, Pentland Science ParkPenicuikUK
| | - Yuying Jiang
- Department of Pharmacology, West China School of PharmacySichuan UniversityChengduChina
| | - Zubing Zhang
- Yiping Medical Science & Technology Development Co. LtdChengduChina
| | | | - Xu Bao
- Department of Pharmacology, West China School of PharmacySichuan UniversityChengduChina
| | - Gary J. Loake
- Green Bioactives Limited, Pentland Science ParkPenicuikUK
- Institute of Molecular Plant Sciences, School of Biological SciencesUniversity of EdinburghEdinburghUK
- Centre for Engineering Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
67
|
Hülpüsch C, Rohayem R, Reiger M, Traidl-Hoffmann C. Exploring the skin microbiome in atopic dermatitis pathogenesis and disease modification. J Allergy Clin Immunol 2024; 154:31-41. [PMID: 38761999 DOI: 10.1016/j.jaci.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 03/04/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
Inflammatory skin diseases such as atopic eczema (atopic dermatitis [AD]) affect children and adults globally. In AD, the skin barrier is impaired on multiple levels. Underlying factors include genetic, chemical, immunologic, and microbial components. Increased skin pH in AD is part of the altered microbial microenvironment that promotes overgrowth of the skin microbiome with Staphylococcus aureus. The secretion of virulence factors, such as toxins and proteases, by S aureus further aggravates the skin barrier deficiency and additionally disrupts the balance of an already skewed immune response. Skin commensal bacteria, however, can inhibit the growth and pathogenicity of S aureus through quorum sensing. Therefore, restoring a healthy skin microbiome could contribute to remission induction in AD. This review discusses direct and indirect approaches to targeting the skin microbiome through modulation of the skin pH; UV treatment; and use of prebiotics, probiotics, and postbiotics. Furthermore, exploratory techniques such as skin microbiome transplantation, ozone therapy, and phage therapy are discussed. Finally, we summarize the latest findings on disease and microbiome modification through targeted immunomodulatory systemic treatments and biologics. We believe that targeting the skin microbiome should be considered a crucial component of successful AD treatment in the future.
Collapse
Affiliation(s)
- Claudia Hülpüsch
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland
| | - Robin Rohayem
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; Dermatology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Chair of Environmental Medicine, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
68
|
Salamat S, Jahan-Mihan A, Gharibvand L, Reza Tabandeh M, Mansoori A. Multi-species synbiotic supplementation increased fecal short chain fatty acids and anti-inflammatory cytokine interleukin-10 in adult men with dyslipidemia; A randomized, double-blind, clinical trial. Cytokine 2024; 179:156608. [PMID: 38631185 DOI: 10.1016/j.cyto.2024.156608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Mounting evidence revealed that an imbalance of Gut Microbiota (GM) leads to metabolic disorders. Synbiotics through regulation of GM composition can be an effective intervention in the management of metabolic diseases. This study aimed to investigate the effects of multi-species synbiotic supplementation on serum interleukin10 (IL-10) and fecal Short Chain Fatty Acids (SCFAs) in patients with dyslipidemia. METHODS In this double-blind, randomized, placebo-controlled clinical trial, fifty-six adult men with dyslipidemia were randomly allocated to intervention and control groups and received either synbiotic or placebo powder twice a day for 12 weeks. Each synbiotic sachet contained 6 species of probiotic microorganisms with a total dose of 3 × 1010 Colony Forming Unit (CFU) and 5 gr inulin and Fructooligosaccharide (FOS) as prebiotics. Blood and stool samples were collected at the baseline and end of the study. Dietary intake, physical activity, anthropometric measurements, serum IL-10, and fecal SCFAs were assessed before and after the intervention. RESULT There were no significant differences between the baseline characteristics of patients in the two groups. Serum IL-10 was increased in the synbiotic group (p < 0.0001). Moreover, synbiotic supplementation increased fecal concentration of acetate (p < 0.0001), butyrate (p = 0.043), propionate (p < 0.0001), and valerate (p < 0.026). A significant positive correlation was observed between the changes in fecal butyrate level and serum IL-10 concentration in the control group (r = 0.48, p = 0.01). CONCLUSIONS A Twelve-week synbiotic supplementation increased fecal SCFAs and improved inflammation in adult men with dyslipidemia.
Collapse
Affiliation(s)
- Shekoufeh Salamat
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Jahan-Mihan
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, USA
| | - Lida Gharibvand
- Loma Linda University School of Allied Health Professions, Loma Linda, CA, USA
| | - Mohammad Reza Tabandeh
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Iran; Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Iran
| | - Anahita Mansoori
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
69
|
Bezemer GFG, Diks MAP, Mortaz E, van Ark I, van Bergenhenegouwen J, Kraneveld AD, Folkerts G, Garssen J. A synbiotic mixture of Bifidobacterium breve M16-V, oligosaccharides and pectin, enhances Short Chain Fatty Acid production and improves lung health in a preclinical model for pulmonary neutrophilia. Front Nutr 2024; 11:1371064. [PMID: 39006103 PMCID: PMC11239554 DOI: 10.3389/fnut.2024.1371064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/15/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction Pulmonary neutrophilia is a hallmark of numerous airway diseases including Chronic Obstructive Pulmonary Disease (COPD), Neutrophilic asthma, Acute Lung Injury (ALI), Acute Respiratory Distress Syndrome (ARDS) and COVID-19. The aim of the current study was to investigate the effect of dietary interventions on lung health in context of pulmonary neutrophilia. Methods Male BALB/cByJ mice received 7 intra-nasal doses of either a vehicle or lipopolysaccharides (LPS). To study the effect of nutritional interventions they received 16 intra-gastric doses of either a vehicle (PBS) or the following supplements (1) probiotic Bifidobacterium breve (B. breve) M16-V; (2) a prebiotic fiber mixture of short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides, and low-viscosity pectin in a 9:1:2 ratio (scGOS/lcFOS/lvPectin); and (3) A synbiotic combination B. breve M16-V and scGOS/lcFOS/lvPectin. Parameters for lung health included lung function, lung morphology and lung inflammation. Parameters for systemic immunomodulation included levels of fecal short chain fatty acids and regulatory T cells. Results The synbiotic supplement protected against the LPS induced decline in lung function (35% improved lung resistance at baseline p = 0.0002 and 25% at peak challenge, p = 0.0002), provided a significant relief from pulmonary neutrophilia (40.7% less neutrophils, p < 0.01) and improved the pulmonary neutrophil-to-lymphocyte ratio (NLR) by 55.3% (p = 0.0033). Supplements did not impact lung morphology in this specific experiment. LPS applied to the upper airways induced less fecal SCFAs production compared to mice that received PBS. The production of acetic acid between day -5 and day 16 was increased in all unchallenged mice (PBS-PBS p = 0.0003; PBS-Pro p < 0.0001; PBS-Pre, p = 0.0045; PBS-Syn, p = 0.0005) which upon LPS challenge was only observed in mice that received the synbiotic mixture of B. breve M16-V and GOS:FOS:lvPectin (p = 0.0003). A moderate correlation was found for butyric acid and lung function parameters and a weak correlation was found between acetic acid, butyric acid and propionic acid concentrations and NLR. Conclusion This study suggests bidirectional gut lung cross-talk in a mouse model for pulmonary neutrophilia. Neutrophilic lung inflammation coexisted with attenuated levels of fecal SCFA. The beneficial effects of the synbiotic mixture of B. breve M16-V and GOS:FOS:lvPectin on lung health associated with enhanced levels of SCFAs.
Collapse
Affiliation(s)
- Gillina F G Bezemer
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Impact Station, Hilversum, Netherlands
| | - Mara A P Diks
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Esmaeil Mortaz
- Department of Microbiology & Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Respiratory Immunology Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ingrid van Ark
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone, Nutricia Research BV, Immunology, Utrecht, Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
- Danone, Nutricia Research BV, Immunology, Utrecht, Netherlands
| |
Collapse
|
70
|
Benvenuti L, Di Salvo C, Bellini G, Seguella L, Rettura F, Esposito G, Antonioli L, Ceravolo R, Bernardini N, Pellegrini C, Fornai M. Gut-directed therapy in Parkinson's disease. Front Pharmacol 2024; 15:1407925. [PMID: 38974034 PMCID: PMC11224490 DOI: 10.3389/fphar.2024.1407925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/17/2024] [Indexed: 07/09/2024] Open
Abstract
Parkinson's disease (PD) is a common and slow-progressing neurodegenerative disorder characterized by motor and non-motor symptoms, including gastrointestinal (GI) dysfunctions. Over the last years, the microbiota-gut-brain (MGB) axis is emerging as a bacterial-neuro-immune ascending pathway that contributes to the progression of PD. Indeed, PD patients are characterized by changes in gut microbiota composition, alterations of intestinal epithelial barrier (IEB) and enteric neurogenic/inflammatory responses that, besides determining intestinal disturbances, contribute to brain pathology. In this context, despite the causal relationship between gut dysbiosis, impaired MGB axis and PD remains to be elucidated, emerging evidence shows that MGB axis modulation can represent a suitable therapeutical strategy for the treatment of PD. This review provides an overview of the available knowledge about the beneficial effects of gut-directed therapies, including dietary interventions, prebiotics, probiotics, synbiotics and fecal microbiota transplantation (FMT), in both PD patients and animal models. In this context, particular attention has been devoted to the mechanisms by which the modulation of MGB axis could halt or slow down PD pathology and, most importantly, how these approaches can be included in the clinical practice.
Collapse
Affiliation(s)
- Laura Benvenuti
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Clelia Di Salvo
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Bellini
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Francesco Rettura
- Unit of Gastroenterology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nunzia Bernardini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carolina Pellegrini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
71
|
Martemucci G, Khalil M, Di Luca A, Abdallah H, D’Alessandro AG. Comprehensive Strategies for Metabolic Syndrome: How Nutrition, Dietary Polyphenols, Physical Activity, and Lifestyle Modifications Address Diabesity, Cardiovascular Diseases, and Neurodegenerative Conditions. Metabolites 2024; 14:327. [PMID: 38921462 PMCID: PMC11206163 DOI: 10.3390/metabo14060327] [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: 04/21/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Several hallmarks of metabolic syndrome, such as dysregulation in the glucose and lipid metabolism, endothelial dysfunction, insulin resistance, low-to-medium systemic inflammation, and intestinal microbiota dysbiosis, represent a pathological bridge between metabolic syndrome and diabesity, cardiovascular, and neurodegenerative disorders. This review aims to highlight some therapeutic strategies against metabolic syndrome involving integrative approaches to improve lifestyle and daily diet. The beneficial effects of foods containing antioxidant polyphenols, intestinal microbiota control, and physical activity were also considered. We comprehensively examined a large body of published articles involving basic, animal, and human studie, as well as recent guidelines. As a result, dietary polyphenols from natural plant-based antioxidants and adherence to the Mediterranean diet, along with physical exercise, are promising complementary therapies to delay or prevent the onset of metabolic syndrome and counteract diabesity and cardiovascular diseases, as well as to protect against neurodegenerative disorders and cognitive decline. Modulation of the intestinal microbiota reduces the risks associated with MS, improves diabetes and cardiovascular diseases (CVD), and exerts neuroprotective action. Despite several studies, the estimation of dietary polyphenol intake is inconclusive and requires further evidence. Lifestyle interventions involving physical activity and reduced calorie intake can improve metabolic outcomes.
Collapse
Affiliation(s)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | - Alessio Di Luca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (A.D.L.); (A.G.D.)
| | - Hala Abdallah
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70121 Bari, Italy;
| | | |
Collapse
|
72
|
Kulshreshtha S. Mushroom as Prebiotics: a Sustainable Approach for Healthcare. Probiotics Antimicrob Proteins 2024; 16:699-712. [PMID: 37776487 DOI: 10.1007/s12602-023-10164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 10/02/2023]
Abstract
Mushrooms are considered as sustainable foods as they require less effort and can be cultivated on different agro-industrial wastes. Besides, these possess many nutraceuticals for providing health benefits along with supplementing nutrition. The mushrooms are also used as prebiotics for their ability to support beneficial microbes in the gut and inhibit the growth of pathogens. Furthermore, these remain undigested in the upper gut and reach the intestine to replenish the gut microbiota. The mushrooms boost health by inhibiting the binding of pathogenic bacteria, by promoting the growth of specific gut microbiota, producing short chain fatty acids, and regulating lipid metabolism and cancer. Research has been initiated in the commercial formulation of various products such as yogurt and symbiotic capsules. This paper sheds light on health-promoting effect, disease controlling, and regulating effect of mushroom prebiotics. This paper also presented a glimpse of commercialization of mushroom prebiotics. In the future, proper standardization of mushroom-based prebiotic formulations will be available to boost human health.
Collapse
Affiliation(s)
- Shweta Kulshreshtha
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India.
| |
Collapse
|
73
|
Corriero A, Giglio M, Soloperto R, Inchingolo F, Varrassi G, Puntillo F. Microbial Symphony: Exploring the Role of the Gut in Osteoarthritis-Related Pain. A Narrative Review. Pain Ther 2024; 13:409-433. [PMID: 38678155 PMCID: PMC11111653 DOI: 10.1007/s40122-024-00602-9] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/29/2024] Open
Abstract
One of the most common musculoskeletal disorders, osteoarthritis (OA), causes worldwide disability, morbidity, and poor quality of life by degenerating articular cartilage, modifying subchondral bone, and inflaming synovial membranes. OA pathogenesis pathways must be understood to generate new preventative and disease-modifying therapies. In recent years, it has been acknowledged that gut microbiota (GM) can significantly contribute to the development of OA. Dysbiosis of GM can disrupt the "symphony" between the host and the GM, leading to a host immunological response that activates the "gut-joint" axis, ultimately worsening OA. This narrative review summarizes research supporting the "gut-joint axis" hypothesis, focusing on the interactions between GM and the immune system in its two main components, innate and adaptive immunity. Furthermore, the pathophysiological sequence of events that link GM imbalance to OA and OA-related pain is broken down and further investigated. We also suggest that diet and prebiotics, probiotics, nutraceuticals, exercise, and fecal microbiota transplantation could improve OA management and represent a new potential therapeutic tool in the light of the scarce panorama of disease-modifying osteoarthritis drugs (DMOADs). Future research is needed to elucidate these complex interactions, prioritizing how a particular change in GM, i.e., a rise or a drop of a specific bacterial strain, correlates with a certain OA subset to pinpoint the associated signaling pathway that leads to OA.
Collapse
Affiliation(s)
- Alberto Corriero
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy.
| | - Mariateresa Giglio
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy
| | - Rossana Soloperto
- Department of Intensive Care, Brussels' University Hospital (HUB), Rue de Lennik 808, 1070, Brussels, Belgium
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124, Bari, Italy
| | | | - Filomena Puntillo
- Department of Interdisciplinary Medicine - ICU Section, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
74
|
Dias TG, Rodrigues LDS, Farias JR, Pereira ALF, Ferreira AGN, Neto MS, Dutra RP, Reis AS, Guerra RNM, Monteiro-Neto V, Maciel MCG. Immunomodulatory Activity of Probiotics in Models of Bacterial Infections. Probiotics Antimicrob Proteins 2024; 16:862-874. [PMID: 37191780 DOI: 10.1007/s12602-023-10090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
As resistance to conventional antibiotics among bacteria continues to increase, researchers are increasingly focusing on alternative strategies for preventing and treating bacterial infections, one of which is microbiota modulation. The objective of this review is to analyze the scientific literature on the immunomodulatory effects of probiotics in bacterial infections. This is an integrative review of the literature based on systematic steps, with searches performed in the databases Medline, PubMed, Scopus, Embase, and ScienceDirect. The most prevalent bacterial genera used to evaluate infectious processes were Salmonella, Escherichia, Klebsiella, and Streptococcus. Lactobacillus was the most commonly used probiotic genus, with Lactobacillus delbrueckii subsp. bulgaricus is the most frequently used species. In most studies, prophylactic treatment with concentrations of probiotics equal to or greater than 8 log CFU/mL was chosen. However, there was considerable heterogeneity in terms of effective treatment duration, indicating that the results cannot be generalized across all studies. This review found that probiotics interact with the immune system through different mechanisms and have a positive effect on preventing different types of bacterial infections.
Collapse
Affiliation(s)
- Tatielle Gomes Dias
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | | | - Josivan Regis Farias
- Graduate Program in Health Sciences, Federal University of Maranhão, São Luís, Maranhão, Brazil
| | - Ana Lúcia Fernandes Pereira
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | - Adriana Gomes Nogueira Ferreira
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | - Marcelino Santos Neto
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | - Richard Pereira Dutra
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | - Aramys Silva Reis
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil
| | - Rosane Nassar Meireles Guerra
- Graduate Program in Health Sciences, Federal University of Maranhão, São Luís, Maranhão, Brazil
- Department of Pathology, Federal University of Maranhão, São Luís, Maranhão, Brazil
| | | | - Márcia Cristina Gonçalves Maciel
- Graduate Program in Health and Technology, Center for Sciences of Imperatriz, Federal University of Maranhão, Maranhão, Brazil.
- Department of Cell Biology, University of Brasília, Brasília, Distrito Federal, Brazil.
| |
Collapse
|
75
|
Biniszewska O, Jacenik D, Tarasiuk A, Fichna J. Current and future pharmacotherapies for the management of constipation-predominant irritable bowel syndrome. Expert Opin Pharmacother 2024; 25:1039-1049. [PMID: 38856704 DOI: 10.1080/14656566.2024.2366993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common gastrointestinal disorder affecting 9-23% of the world's population, with a higher prevalence among women. IBS is a complex disorder influenced by psychosocial, physiological, and genetic factors, exacerbated by stress. AREAS COVERED Research confirms that the most common subtype of IBS is IBS-C. Therefore, new therapies are being developed to speed up bowel movement and reduce constipation, with drugs such as linaclotide, plecanatide, lubiprostone, or tegaserod available to reduce IBS-C symptoms. In addition, patients' condition is improved by foods rich in fiber and low in FODMAP and the use of biotics. EXPERT OPINION The topic is of great importance due to the growing number of patients suffering from IBS-C and its significant impact on quality of life. Current clinical trials of new therapeutic options are not too successful, and it seems that one of the plausible treatment options could be the multi-drug cocktail with some, or perhaps even all its ingredients emerging from drug re-purposing. Another important path that needs to be explored further in IBS-C patients is the adjustment of dietary habits and/or introduction of dietary or nutritional intervention.
Collapse
Affiliation(s)
- Olga Biniszewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
76
|
Babu A, Devi Rajeswari V, Ganesh V, Das S, Dhanasekaran S, Usha Rani G, Ramanathan G. Gut Microbiome and Polycystic Ovary Syndrome: Interplay of Associated Microbial-Metabolite Pathways and Therapeutic Strategies. Reprod Sci 2024; 31:1508-1520. [PMID: 38228976 DOI: 10.1007/s43032-023-01450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a multifaceted disease with an intricate etiology affecting reproductive-aged women. Despite attempts to unravel the pathophysiology, the molecular mechanism of PCOS remains unknown. There are no effective or suitable therapeutic strategies available to ameliorate PCOS; however, the symptoms can be managed. In recent years, a strong association has been found between the gut microbiome and PCOS, leading to the formulation of novel ideas on the genesis and pathological processes of PCOS. Further, gut microbiome dysbiosis involving microbial metabolites may trigger PCOS symptoms via many mechanistic pathways including those associated with carbohydrates, short-chain fatty acids, lipopolysaccharides, bile acids, and gut-brain axis. We present the mechanistic pathways of PCOS-related microbial metabolites and therapeutic opportunities available to treat PCOS, such as prebiotics, probiotics, and fecal microbiota therapy. In addition, the current review highlights the emerging treatment strategies available to alleviate the symptoms of PCOS.
Collapse
Affiliation(s)
- Achsha Babu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - V Devi Rajeswari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - V Ganesh
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Soumik Das
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sivaraman Dhanasekaran
- Pandit Deendayal Energy University, Knowledge Corridor, Raisan Village, PDPU Road, Gandhinagar, Gujarat, 382426, India
| | - G Usha Rani
- Department of Obstetrics And Gynecology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
77
|
Shang Z, Pai L, Patil S. Unveiling the dynamics of gut microbial interactions: a review of dietary impact and precision nutrition in gastrointestinal health. Front Nutr 2024; 11:1395664. [PMID: 38873568 PMCID: PMC11169903 DOI: 10.3389/fnut.2024.1395664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/21/2024] [Indexed: 06/15/2024] Open
Abstract
The human microbiome, a dynamic ecosystem within the gastrointestinal tract, plays a pivotal role in shaping overall health. This review delves into six interconnected sections, unraveling the intricate relationship between diet, gut microbiota, and their profound impact on human health. The dance of nutrients in the gut orchestrates a complex symphony, influencing digestive processes and susceptibility to gastrointestinal disorders. Emphasizing the bidirectional communication between the gut and the brain, the Brain-Gut Axis section highlights the crucial role of dietary choices in physical, mental, and emotional well-being. Autoimmune diseases, particularly those manifesting in the gastrointestinal tract, reveal the delicate balance disrupted by gut microbiome imbalances. Strategies for reconciling gut microbes through diets, precision nutrition, and clinical indications showcase promising avenues for managing gastrointestinal distress and revolutionizing healthcare. From the Low-FODMAP diet to neuro-gut interventions, these strategies provide a holistic understanding of the gut's dynamic world. Precision nutrition, as a groundbreaking discipline, holds transformative potential by tailoring dietary recommendations to individual gut microbiota compositions, reshaping the landscape of gastrointestinal health. Recent advancements in clinical indications, including exact probiotics, fecal microbiota transplantation, and neuro-gut interventions, signify a new era where the gut microbiome actively participates in therapeutic strategies. As the microbiome takes center stage in healthcare, a paradigm shift toward personalized and effective treatments for gastrointestinal disorders emerges, reflecting the symbiotic relationship between the human body and its microbial companions.
Collapse
Affiliation(s)
- Zifang Shang
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou People's Hospital (Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou, China
| | - Liu Pai
- Department of Haematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Sandip Patil
- Department of Haematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
78
|
Sivaprasadan S, Anila KN, Nair K, Mallick S, Biswas L, Valsan A, Praseedom RK, Nair BKG, Sudhindran S. Microbiota and Gut-Liver Axis: An Unbreakable Bond? Curr Microbiol 2024; 81:193. [PMID: 38805045 DOI: 10.1007/s00284-024-03694-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/08/2024] [Indexed: 05/29/2024]
Abstract
The gut microbiota, amounting to approximately 100 trillion (1014) microbes represents a genetic repertoire that is bigger than the human genome itself. Evidence on bidirectional interplay between human and microbial genes is mounting. Microbiota probably play vital roles in diverse aspects of normal human metabolism, such as digestion, immune modulation, and gut endocrine function, as well as in the genesis and progression of many human diseases. Indeed, the gut microbiota has been most closely linked to various chronic ailments affecting the liver, although concrete scientific data are sparse. In this narrative review, we initially discuss the basic epidemiology of gut microbiota and the factors influencing their initial formation in the gut. Subsequently, we delve into the gut-liver axis and the evidence regarding the link between gut microbiota and the genesis or progression of various liver diseases. Finally, we summarise the recent research on plausible ways to modulate the gut microbiota to alter the natural history of liver disease.
Collapse
Affiliation(s)
- Saraswathy Sivaprasadan
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - K N Anila
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Krishnanunni Nair
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Shweta Mallick
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Lalitha Biswas
- Amrita School of Nanosciences and Molecular Medicine, Kochi, India
| | - Arun Valsan
- Department of Hepatology & Gastroenterology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India
| | | | | | - Surendran Sudhindran
- Department of Gastrointestinal Surgery and Solid Organ Transplantation, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, India.
| |
Collapse
|
79
|
Duysburgh C, Govaert M, Guillemet D, Marzorati M. Co-Supplementation of Baobab Fiber and Arabic Gum Synergistically Modulates the In Vitro Human Gut Microbiome Revealing Complementary and Promising Prebiotic Properties. Nutrients 2024; 16:1570. [PMID: 38892504 PMCID: PMC11173755 DOI: 10.3390/nu16111570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Arabic gum, a high molecular weight heteropolysaccharide, is a promising prebiotic candidate as its fermentation occurs more distally in the colon, which is the region where most chronic colonic diseases originate. Baobab fiber could be complementary due to its relatively simple structure, facilitating breakdown in the proximal colon. Therefore, the current study aimed to gain insight into how the human gut microbiota was affected in response to long-term baobab fiber and Arabic gum supplementation when tested individually or as a combination of both, allowing the identification of potential complementary and/or synergetic effects. The validated Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), an in vitro gut model simulating the entire human gastrointestinal tract, was used. The microbial metabolic activity was examined, and quantitative 16S-targeted Illumina sequencing was used to monitor the gut microbial composition. Moreover, the effect on the gut microbial metabolome was quantitatively analyzed. Repeated administration of baobab fiber, Arabic gum, and their combination had a significant effect on the metabolic activity, diversity index, and community composition of the microbiome present in the simulated proximal and distal colon with specific impacts on Bifidobacteriaceae and Faecalibacterium prausnitzii. Despite the lower dosage strategy (2.5 g/day), co-supplementation of both compounds resulted in some specific synergistic prebiotic effects, including a biological activity throughout the entire colon, SCFA synthesis including a synergy on propionate, specifically increasing abundance of Akkermansiaceae and Christensenellaceae in the distal colon region, and enhancing levels of spermidine and other metabolites of interest (such as serotonin and ProBetaine).
Collapse
Affiliation(s)
- Cindy Duysburgh
- ProDigest Bv, Technologiepark 82, 9052 Ghent, Belgium; (C.D.); (M.G.)
| | - Marlies Govaert
- ProDigest Bv, Technologiepark 82, 9052 Ghent, Belgium; (C.D.); (M.G.)
| | | | - Massimo Marzorati
- ProDigest Bv, Technologiepark 82, 9052 Ghent, Belgium; (C.D.); (M.G.)
- Center of Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| |
Collapse
|
80
|
Cardoso BB, Amorim C, Franco-Duarte R, Alves JI, Barbosa SG, Silvério SC, Rodrigues LR. Epilactose as a Promising Butyrate-Promoter Prebiotic via Microbiota Modulation. Life (Basel) 2024; 14:643. [PMID: 38792663 PMCID: PMC11123345 DOI: 10.3390/life14050643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Epilactose is a disaccharide composed of galactose and mannose, and it is currently considered an "under development" prebiotic. In this study, we described the prebiotic potential of epilactose by in vitro fermentation using human fecal inocula from individuals following a Mediterranean diet (DM) or a Vegan diet (DV). The prebiotic effect of epilactose was also compared with lactulose and raffinose, and interesting correlations were established between metabolites and microbiota modulation. The production of several metabolites (lactate, short-chain fatty acids, and gases) confirmed the prebiotic properties of epilactose. For both donors, the microbiota analysis showed that epilactose significantly stimulated the butyrate-producing bacteria, suggesting that its prebiotic effect could be independent of the donor diet. Butyrate is one of the current golden metabolites due to its benefits for the gut and systemic health. In the presence of epilactose, the production of butyrate was 70- and 63-fold higher for the DM donor, when compared to lactulose and raffinose, respectively. For the DV donor, an increase of 29- and 89-fold in the butyrate production was obtained when compared to lactulose and raffinose, respectively. In conclusion, this study suggests that epilactose holds potential functional properties for human health, especially towards the modulation of butyrate-producing strains.
Collapse
Affiliation(s)
- Beatriz B. Cardoso
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
| | - Cláudia Amorim
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
- LABBELS—Associate Laboratory, Guimarães, 4710-057 Braga, Portugal
| | - Ricardo Franco-Duarte
- CBMA—Centre of Molecular and Environmental Biology, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
| | - Joana I. Alves
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
- LABBELS—Associate Laboratory, Guimarães, 4710-057 Braga, Portugal
| | - Sónia G. Barbosa
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
- LABBELS—Associate Laboratory, Guimarães, 4710-057 Braga, Portugal
| | - Sara C. Silvério
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
- LABBELS—Associate Laboratory, Guimarães, 4710-057 Braga, Portugal
| | - Lígia R. Rodrigues
- CEB—Centre of Biological Engineering, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (B.B.C.); (C.A.); (J.I.A.); (S.G.B.); (S.C.S.)
- LABBELS—Associate Laboratory, Guimarães, 4710-057 Braga, Portugal
| |
Collapse
|
81
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
82
|
Dahiya P, Kumari S, Behl M, Kashyap A, Kumari D, Thakur K, Devi M, Kumari N, Kaushik N, Walia A, Bhatt AK, Bhatia RK. Guardians of the Gut: Harnessing the Power of Probiotic Microbiota and Their Exopolysaccharides to Mitigate Heavy Metal Toxicity in Human for Better Health. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10281-9. [PMID: 38733461 DOI: 10.1007/s12602-024-10281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Abstract
Heavy metal pollution is a significant global health concern, posing risks to both the environment and human health. Exposure to heavy metals happens through various channels like contaminated water, food, air, and workplaces, resulting in severe health implications. Heavy metals also disrupt the gut's microbial balance, leading to dysbiosis characterized by a decrease in beneficial microorganisms and proliferation in harmful ones, ultimately exacerbating health problems. Probiotic microorganisms have demonstrated their ability to adsorb and sequester heavy metals, while their exopolysaccharides (EPS) exhibit chelating properties, aiding in mitigating heavy metal toxicity. These beneficial microorganisms aid in restoring gut integrity through processes like biosorption, bioaccumulation, and biotransformation of heavy metals. Incorporating probiotic strains with high affinity for heavy metals into functional foods and supplements presents a practical approach to mitigating heavy metal toxicity while enhancing gut health. Utilizing probiotic microbiota and their exopolysaccharides to address heavy metal toxicity offers a novel method for improving human health through modulation of the gut microbiome. By combining probiotics and exopolysaccharides, a distinctive strategy emerges for mitigating heavy metal toxicity, highlighting promising avenues for therapeutic interventions and health improvements. Further exploration in this domain could lead to groundbreaking therapies and preventive measures, underscoring probiotic microbiota and exopolysaccharides as natural and environmentally friendly solutions to heavy metal toxicity. This, in turn, could enhance public health by safeguarding the gut from environmental contaminants.
Collapse
Affiliation(s)
- Pushpak Dahiya
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Sangeeta Kumari
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Manya Behl
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Aakash Kashyap
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Deeksha Kumari
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Kalpana Thakur
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Mamta Devi
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Neelam Kumari
- Department of Biosciences, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Neelam Kaushik
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Abhishek Walia
- Department of Microbiology, College of Basic Sciences, CSK HPKV, Palampur, HP, 176062, India
| | - Arvind Kumar Bhatt
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Ravi Kant Bhatia
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India.
| |
Collapse
|
83
|
Gyriki D, Nikolaidis C, Stavropoulou E, Bezirtzoglou I, Tsigalou C, Vradelis S, Bezirtzoglou E. Exploring the Gut Microbiome's Role in Inflammatory Bowel Disease: Insights and Interventions. J Pers Med 2024; 14:507. [PMID: 38793089 PMCID: PMC11122163 DOI: 10.3390/jpm14050507] [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: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Inflammatory Bowel Disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a chronic and relapsing inflammatory condition of the intestine that significantly impairs quality of life and imposes a heavy burden on healthcare systems globally. While the exact etiology of IBD is unclear, it is influenced by genetic, environmental, immunological, and microbial factors. Recent advances highlight the gut microbiome's pivotal role in IBD pathogenesis. The microbial dysbiosis characteristic of IBD, marked by a decline in beneficial bacteria and an increase in pathogenic microbes, suggests a profound connection between microbial imbalance and disease mechanisms. This review explores diagnostic approaches to IBD that integrate clinical assessment with advanced microbiological analyses, highlighting the potential of microbiome profiling as a non-invasive diagnostic tool. In addition, it evaluates conventional and emerging treatments and discusses microbiome-targeted intervention prospects, such as probiotics, symbiotics, and faecal microbiota transplantation. The necessity for future research to establish their efficacy and safety is emphasised.
Collapse
Affiliation(s)
- Despoina Gyriki
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.T.); (S.V.); (E.B.)
- Internal Medicine Department, Vostaneio-General Hospital of Mytilene, 81100 Mytilene, Greece;
| | - Christos Nikolaidis
- Internal Medicine Department, Vostaneio-General Hospital of Mytilene, 81100 Mytilene, Greece;
| | - Elisavet Stavropoulou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.T.); (S.V.); (E.B.)
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | | | - Christina Tsigalou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.T.); (S.V.); (E.B.)
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Stergios Vradelis
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.T.); (S.V.); (E.B.)
- Department of Gastroenterology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Eugenia Bezirtzoglou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (C.T.); (S.V.); (E.B.)
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
84
|
Ní Chonnacháin C, Feeney EL, Gollogly C, Shields DC, Loscher CE, Cotter PD, Noronha N, Stack R, Doherty GA, Gibney ER. The effects of dairy on the gut microbiome and symptoms in gastrointestinal disease cohorts: a systematic review. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e5. [PMID: 39290657 PMCID: PMC11406376 DOI: 10.1017/gmb.2024.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/13/2024] [Accepted: 04/10/2024] [Indexed: 09/19/2024]
Abstract
Bovine dairy foods provide several essential nutrients. Fermented bovine dairy foods contain additional compounds, increasing their potential to benefit gastrointestinal health. This review explores the effects of dairy consumption on the gut microbiome and symptoms in gastrointestinal disease cohorts. Human subjects with common gastrointestinal diseases (functional gastrointestinal disorders and inflammatory bowel disease) or associated symptoms, and equivalent animal models were included. A systematic literature search was performed using PubMed, Embase and Web of Science. The search yielded 3014 studies in total, with 26 meeting inclusion criteria, including 15 human studies (1550 participants) and 11 animal studies (627 subjects). All test foods were fermented bovine dairy products, primarily fermented milk and yogurt. Six studies reported increases in gastrointestinal bacterial alpha diversity, with nine studies reporting increases in relative Lactobacillus and Bifidobacterium abundance. Six studies reported increases in beneficial short-chain fatty acids, while three reported decreases. Gastrointestinal symptoms, specifically gut comfort and defecation frequency, improved in 14 human studies. Five animal studies demonstrated reduced colonic damage and improved healing. This review shows fermented bovine dairy consumption may improve gut microbial characteristics and gastrointestinal symptoms in gastrointestinal disease cohorts. Further human intervention studies are needed, expanding test foods and capturing non-self-reported gastrointestinal measures.
Collapse
Affiliation(s)
- Clíona Ní Chonnacháin
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Emma L Feeney
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Clare Gollogly
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Denis C Shields
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Christine E Loscher
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Paul D Cotter
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Department of Food Biosciences, Teagasc Food Research Centre, APC Microbiome Ireland and VistaMilk, Dublin, Ireland
| | - Nessa Noronha
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Institute of Food and Health, University College Dublin, Dublin, Ireland
| | - Roisin Stack
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Glen A Doherty
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Eileen R Gibney
- Food for Health Ireland, University College Dublin, Dublin, Ireland
- Institute of Food and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
85
|
Colombo R, Moretto G, Pellicorio V, Papetti A. Globe Artichoke ( Cynara scolymus L.) By-Products in Food Applications: Functional and Biological Properties. Foods 2024; 13:1427. [PMID: 38790727 PMCID: PMC11119529 DOI: 10.3390/foods13101427] [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: 04/10/2024] [Revised: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Globe artichoke (Cynara cardunculus var. scolymus L.) is widely cultivated in the Mediterranean area and Italy is one of the largest producers. A great issue is represented by its high amount of by-product, mainly consisting of external bracts and stems, but also of residual leaves, stalks, roots, and seeds. Artichoke by-products are rich in nutrients (carbohydrates and proteins) and bioactive compounds (polyphenols and terpenes) and represent potential ingredients for foodstuffs, functional foods, and food supplements, due to their functional and biological properties. In fact, artichoke by-products' components exhibit many beneficial effects, such as dyspeptic, prebiotic, antioxidant, anti-inflammatory, antiglycative, antimicrobial, anticarcinogenic, and hypolipidemic properties. Therefore, they can be considered potential food ingredients useful in reducing the risk of developing metabolic and age-related disorders. This work summarizes the economic and environmental impact of the recovery and valorization of artichoke by-products, focusing on rheological, physical, and biological properties of the different components present in each by-product and their different food applications.
Collapse
Affiliation(s)
- Raffaella Colombo
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (R.C.); (G.M.); (V.P.)
| | - Giulia Moretto
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (R.C.); (G.M.); (V.P.)
| | - Vanessa Pellicorio
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (R.C.); (G.M.); (V.P.)
| | - Adele Papetti
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy; (R.C.); (G.M.); (V.P.)
- C.S.G.I., University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
86
|
van de Put B, de Bruijn WJ, Schols HA. Structural Characterization of Disaccharides Using Cyclic Ion Mobility Spectrometry and Monosaccharide Standards. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1012-1020. [PMID: 38634722 PMCID: PMC11066964 DOI: 10.1021/jasms.4c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
To understand the mode of action of bioactive oligosaccharides, such as prebiotics, in-depth knowledge about all structural features, including monosaccharide composition, linkage type, and anomeric configuration, is necessary. Current analytical techniques provide limited information about structural features within complex mixtures unless preceded by extensive purification. In this study, we propose an approach employing cyclic ion mobility spectrometry (cIMS) for the in-depth characterization of oligosaccharides, here demonstrated for disaccharides. We were able to separate galactose and glucose anomers by exploiting the high ion mobility resolution of cIMS. Using the obtained monosaccharide mobilograms as references, we determined the composition and anomeric configuration of 4β-galactobiose by studying the monosaccharide fragments generated by collision-induced dissociation (CID) before the ion mobility separation. Drift times and individual MS2 spectra of partially resolved reducing-end anomers of 4β-galactobiose, 4β-galactosylglucose (lactose), and 4β-glucosylglucose (cellobiose) were obtained by deconvolution using CID fragmentation induced in the transfer region between the cIMS cell and TOF analyzer. The composition and anomeric configuration of the reducing end anomers of these disaccharides were identified using cIMS2 approaches, where first each anomer was isolated using cIMS and individually fragmented, and the monosaccharide fragments were again separated by cIMS for comparison with monosaccharide standards. With these results we demonstrate the promising application of cIMS for the structural characterization of isomeric oligosaccharides.
Collapse
Affiliation(s)
- Bram van de Put
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| | - Wouter J.C. de Bruijn
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| | - Henk A. Schols
- Laboratory of Food Chemistry, Wageningen University, Bornse Weilanden 9, 6708, WG Wageningen, The Netherlands
| |
Collapse
|
87
|
McCoubrey LE, Ferraro F, Seegobin N, Verin J, Alfassam HA, Awad A, Marzorati M, Verstrepen L, Ghyselinck J, De Munck J, De Medts J, Steppe E, De Vleeschhauwer V, De Rocker G, Droesbeke A, De Rijck M, Vanthoor S, Moens F, Siepmann J, Siepmann F, Gaisford S, Orlu M, Basit AW. Poly(D,l-lactide-co-glycolide) particles are metabolised by the gut microbiome and elevate short chain fatty acids. J Control Release 2024; 369:163-178. [PMID: 38521168 DOI: 10.1016/j.jconrel.2024.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/17/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
The production of short chain fatty acids (SCFAs) by the colonic microbiome has numerous benefits for human health, including maintenance of epithelial barrier function, suppression of colitis, and protection against carcinogenesis. Despite the therapeutic potential, there is currently no optimal approach for elevating the colonic microbiome's synthesis of SCFAs. In this study, poly(D,l-lactide-co-glycolide) (PLGA) was investigated for this application, as it was hypothesised that the colonic microbiota would metabolise PLGA to its lactate monomers, which would promote the resident microbiota's synthesis of SCFAs. Two grades of spray dried PLGA, alongside a lactate bolus control, were screened in an advanced model of the human colon, known as the M-SHIME® system. Whilst the high molecular weight (Mw) grade of PLGA was stable in the presence of the microbiota sourced from three healthy humans, the low Mw PLGA (PLGA 2) was found to be metabolised. This microbial degradation led to sustained release of lactate over 48 h and increased concentrations of the SCFAs propionate and butyrate. Further, microbial synthesis of harmful ammonium was significantly reduced compared to untreated controls. Interestingly, both types of PLGA were found to influence the composition of the luminal and mucosal microbiota in a donor-specific manner. An in vitro model of an inflamed colonic epithelium also showed the polymer to affect the expression of pro- and anti-inflammatory markers, such as interleukins 8 and 10. The findings of this study reveal PLGA's sensitivity to enzymatic metabolism in the gut, which could be harnessed for therapeutic elevation of colonic SCFAs.
Collapse
Affiliation(s)
- Laura E McCoubrey
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Fabiana Ferraro
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - Nidhi Seegobin
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Jérémy Verin
- Univ. Lille, Inserm, CHU Lille, U1008, F-59000 Lille, France
| | - Haya A Alfassam
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), 114422 Riyadh, Saudi Arabia
| | - Atheer Awad
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom; Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, United Kingdom
| | | | | | | | | | | | - Evi Steppe
- ProDigest BVB, Technologiepark 73, 9052 Ghent, Belgium
| | | | | | | | | | - Sara Vanthoor
- ProDigest BVB, Technologiepark 73, 9052 Ghent, Belgium
| | | | | | | | - Simon Gaisford
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Mine Orlu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom.
| |
Collapse
|
88
|
Mahroof M, Dar RA, Nazir R, Ali MN, Ganai BA. Valorization of rice straw and vascular aquatic weeds for sustainable prebiotic hemicellulosic autohydrolysate production: Extraction, characterization and fermentability. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:35744-35759. [PMID: 38744764 DOI: 10.1007/s11356-024-33611-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
This study describes the extraction and characterization of the hemicellulosic autohydrolysates (HAHs) derived from rice straw (RS) and vascular aquatic weeds like Typha angustifolia (TA) and Ceretophyllum demersum (CD). It further explores their capacity to sustain the proliferation of selected lactic acid bacteria (i.e., prebiotic activity) isolated from milk samples. To fractionate HAH from RS, TA and CD hot water extraction (HWE) method was used and RS, TA, and CD biomasses yielded 6.8, 4.99 and 2.98% of HAH corresponding to the hemicellulose extraction efficiencies of 26.15 ± 0.8%, 23.76 ± 0.6%, and 18.62 ± 0.4% respectively. The chemical characterization of HAH concentrates through HPLC showed that they comprised galactose, arabinose, xylose and glucose. The total phenol content of the RS, TA and CD-derived HAH concentrates were 37.53, 56.78 and 48.08 mg GAE/g. The five lactic acid bacteria (LAB) isolates Q1B, Q2A, Q3B, G1C and G2B selected for prebiotic activity assays generated mixed responses with the highest growth in RS-HAH for Q2A and the least in TA-HAH for Q3B. Further, the isolates Q2A, Q3B, G1C, and G2B, which showed the highest growth performance, were identified through MALDI-TOF and 16S rRNA sequencing as Lactobacillus brevis. All the tested LAB isolates showed diauxic growth in crude HAH preparations to maximize the utilization of carbon resources for their proliferation. This suggests that the selected LAB isolates are efficient degraders of hemicellulosic sugars. This paves the way for the valorization of lignocellulosic biomass to produce prebiotic hemicellulosic autohydrolysate and consequently enhances environmental sustainability by improving resource efficiency.
Collapse
Affiliation(s)
- Mawish Mahroof
- Centre of Research for Development (CORD), University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Rouf Ahmad Dar
- Centre of Research for Development (CORD), University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India.
| | - Ruqeya Nazir
- Centre of Research for Development (CORD), University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Md Niamat Ali
- Centre of Research for Development (CORD), University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Hazratbal, Srinagar, Jammu and Kashmir, 190006, India
| |
Collapse
|
89
|
Solberg BS, Kvalvik LG, Instanes JT, Hartman CA, Klungsøyr K, Li L, Larsson H, Magnus P, Njølstad PR, Johansson S, Andreassen OA, Bakken NR, Bekkhus M, Austerberry C, Smajlagic D, Havdahl A, Corfield EC, Haavik J, Gjestad R, Zayats T. Maternal Fiber Intake During Pregnancy and Development of Attention-Deficit/Hyperactivity Disorder Symptoms Across Childhood: The Norwegian Mother, Father, and Child Cohort Study. Biol Psychiatry 2024; 95:839-848. [PMID: 38142720 DOI: 10.1016/j.biopsych.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Epidemiological studies suggest that maternal diet quality during pregnancy may influence the risk of neurodevelopmental disorders in offspring. Here, we investigated associations between maternal intake of dietary fiber and attention-deficit/hyperactivity disorder (ADHD) symptoms in early childhood. METHODS We used longitudinal data of up to 21,852 mother-father-child trios (49.2% female offspring) from MoBa (the Norwegian Mother, Father, and Child Cohort Study). The relationships between maternal fiber intake during pregnancy and offspring ADHD symptoms at ages 3, 5, and 8 years were examined using 1) multivariate regression (overall levels of ADHD symptoms), 2) latent class analysis (subclasses of ADHD symptoms by sex at each age), and 3) latent growth curves (longitudinal change in offspring ADHD symptoms). Covariates were ADHD polygenic scores in child and parents, total energy intake and energy-adjusted sugar intake, parental ages at birth of the child, and sociodemographic factors. RESULTS Higher maternal prenatal fiber intake was associated with lower offspring ADHD symptom scores at all ages (Bage3 = -0.14 [95% CI, -0.18 to -0.10]; Bage5 = -0.14 [95% CI, -0.19 to -0.09]; Bage8 = -0.14 [95% CI, -0.20 to -0.09]). Of the derived low/middle/high subclasses of ADHD symptoms, fiber was associated with lower risk of belonging to the middle subclass for boys and girls and to the high subclass for girls only (middle: odds ratioboys 0.91 [95% CI, 0.86 to 0.97]/odds ratiogirls 0.86 [95% CI, 0.81 to 0.91]; high: odds ratiogirls 0.82 [95% CI, 0.72 to 0.94]). Maternal fiber intake and rate of change in child ADHD symptoms across ages were not associated. CONCLUSIONS Low prenatal maternal fiber intake may increase symptom levels of ADHD in offspring during childhood, independently of genetic predisposition to ADHD, unhealthy dietary exposures, and sociodemographic factors.
Collapse
Affiliation(s)
- Berit Skretting Solberg
- Department of Biomedicine, University of Bergen, Norway; Child and Adolescent Psychiatric Outpatient Unit, Hospital Betanien, Bergen, Norway.
| | | | | | - Catharina A Hartman
- Interdisciplinary Center Psychiatry and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Kari Klungsøyr
- Department of Global Public Health and Primary Care, University of Bergen, Norway; Division of Mental and Physical Health, Norwegian Institute of Public Health, Bergen, Norway
| | - Lin Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål Rasmus Njølstad
- Department of Clinical Science, Mohn Center for Diabetes Precision Medicine, University of Bergen, Bergen, Norway; Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stefan Johansson
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiciton, Oslo University Hospital, Oslo, Norway
| | - Nora Refsum Bakken
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Mona Bekkhus
- Promenta Research Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Chloe Austerberry
- Centre for Family Research, University of Cambridge, Cambridge, United Kingdom; Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom
| | - Dinka Smajlagic
- Promenta Research Centre, Department of Psychology, University of Oslo, Oslo, Norway
| | - Alexandra Havdahl
- Promenta Research Centre, Department of Psychology, University of Oslo, Oslo, Norway; Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway; Nic Waals Institute, Lovisenberg Diakonale Hospital, Oslo, Norway
| | - Elizabeth C Corfield
- Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway; Nic Waals Institute, Lovisenberg Diakonale Hospital, Oslo, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Norway; Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Psychiatry, Research Department, Haukeland University Hospital, Bergen, Norway
| | - Rolf Gjestad
- Department of Psychiatry, Research Department, Haukeland University Hospital, Bergen, Norway; Center for Crisis Psychology, Faculty of Psychology, University of Bergen, Bergen, Norway; Centre for Research and Education in Forensic Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Tetyana Zayats
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
90
|
Zoghi S, Sadeghpour Heravi F, Nikniaz Z, Shirmohamadi M, Moaddab SY, Ebrahimzadeh Leylabadlo H. Gut microbiota and childhood malnutrition: Understanding the link and exploring therapeutic interventions. Eng Life Sci 2024; 24:2300070. [PMID: 38708416 PMCID: PMC11065333 DOI: 10.1002/elsc.202300070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 05/07/2024] Open
Abstract
Childhood malnutrition is a metabolic condition that affects the physical and mental well-being of children and leads to resultant disorders in maturity. The development of childhood malnutrition is influenced by a number of physiological and environmental factors including metabolic stress, infections, diet, genetic variables, and gut microbiota. The imbalanced gut microbiota is one of the main environmental risk factors that significantly influence host physiology and childhood malnutrition progression. In this review, we have evaluated the gut microbiota association with undernutrition and overnutrition in children, and then the quantitative and qualitative significance of gut dysbiosis in order to reveal the impact of gut microbiota modification using probiotics, prebiotics, synbiotics, postbiotics, fecal microbiota transplantation, and engineering biology methods as new therapeutic challenges in the management of disturbed energy homeostasis. Understanding the host-microbiota interaction and the remote regulation of other organs and pathways by gut microbiota can improve the effectiveness of new therapeutic approaches and mitigate the negative consequences of childhood malnutrition.
Collapse
Affiliation(s)
- Sevda Zoghi
- Liver and Gastrointestinal Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | - Masoud Shirmohamadi
- Liver and Gastrointestinal Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | - Seyed Yaghoub Moaddab
- Liver and Gastrointestinal Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | | |
Collapse
|
91
|
Zhong S, Yang J, Huang H. The role of single and mixed biofilms in Clostridioides difficile infection and strategies for prevention and inhibition. Crit Rev Microbiol 2024; 50:285-299. [PMID: 36939635 DOI: 10.1080/1040841x.2023.2189950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/06/2023] [Indexed: 03/21/2023]
Abstract
Clostridioides difficile infection (CDI) is a serious disease with a high recurrence rate. The single and mixed biofilms formed by C. difficile in the gut contribute to the formation of recurrent CDI (rCDI). In parallel, other gut microbes influence the formation and development of C. difficile biofilms, also known as symbiotic biofilms. Interactions between members within the symbiotic biofilm are associated with the worsening or alleviation of CDI. These interactions include effects on C. difficile adhesion and chemotaxis, modulation of LuxS/AI-2 quorum sensing (QS) system activity, promotion of cross-feeding by microbial metabolites, and regulation of intestinal bile acid and pyruvate levels. In the process of C. difficile biofilms control, inhibition of C. difficile initial biofilm formation and killing of C. difficile vegetative cells and spores are the main targets of action. The role of symbiotic biofilms in CDI suggested that targeting interventions of C. difficile-promoting gut microbes could indirectly inhibit the formation of C. difficile mixed biofilms and improved the ultimate therapeutic effect. In summary, this review outlines the mechanisms of C. difficile biofilm formation and summarises the treatment strategies for such single and mixed biofilms, aiming to provide new ideas for the prevention and treatment of CDI.
Collapse
Affiliation(s)
- Saiwei Zhong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
92
|
Murali SK, Mansell TJ. Next generation probiotics: Engineering live biotherapeutics. Biotechnol Adv 2024; 72:108336. [PMID: 38432422 DOI: 10.1016/j.biotechadv.2024.108336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
The population dynamics of the human microbiome have been associated with inflammatory bowel disease, cancer, obesity, autoimmune diseases, and many other human disease states. An emerging paradigm in treatment is the administration of live engineered organisms, also called next-generation probiotics. However, the efficacy of these microbial therapies can be limited by the organism's overall performance in the harsh and nutrient-limited environment of the gut. In this review, we summarize the current state of the art use of bacterial and yeast strains as probiotics, highlight the recent development of genetic tools for engineering new therapeutic functions in these organisms, and report on the latest therapeutic applications of engineered probiotics, including recent clinical trials. We also discuss the supplementation of prebiotics as a method of manipulating the microbiome and improving the overall performance of engineered live biotherapeutics.
Collapse
Affiliation(s)
- Sanjeeva Kumar Murali
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Thomas J Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA; Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
93
|
Paudel D, Nair DVT, Joseph G, Castro R, Tiwari AK, Singh V. Gastrointestinal microbiota-directed nutritional and therapeutic interventions for inflammatory bowel disease: opportunities and challenges. Gastroenterol Rep (Oxf) 2024; 12:goae033. [PMID: 38690290 PMCID: PMC11057942 DOI: 10.1093/gastro/goae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 05/02/2024] Open
Abstract
Evidence-based research has confirmed the role of gastrointestinal microbiota in regulating intestinal inflammation. These data have generated interest in developing microbiota-based therapies for the prevention and management of inflammatory bowel disease (IBD). Despite in-depth understanding of the etiology of IBD, it currently lacks a cure and requires ongoing management. Accumulating data suggest that an aberrant gastrointestinal microbiome, often referred to as dysbiosis, is a significant environmental instigator of IBD. Novel microbiome-targeted interventions including prebiotics, probiotics, fecal microbiota transplant, and small molecule microbiome modulators are being evaluated as therapeutic interventions to attenuate intestinal inflammation by restoring a healthy microbiota composition and function. In this review, the effectiveness and challenges of microbiome-centered interventions that have the potential to alleviate intestinal inflammation and improve clinical outcomes of IBD are explored.
Collapse
Affiliation(s)
- Devendra Paudel
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Divek V T Nair
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Grace Joseph
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Rita Castro
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Amit K Tiwari
- College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
94
|
Zheng Y, Qin C, Wen M, Zhang L, Wang W. The Effects of Food Nutrients and Bioactive Compounds on the Gut Microbiota: A Comprehensive Review. Foods 2024; 13:1345. [PMID: 38731716 PMCID: PMC11083588 DOI: 10.3390/foods13091345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/06/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
It is now widely recognized that gut microbiota plays a critical role not only in the development and progression of diseases, but also in its susceptibility to dietary patterns, food composition, and nutritional intake. In this comprehensive review, we have compiled the latest findings on the effects of food nutrients and bioactive compounds on the gut microbiota. The research indicates that certain components, such as unsaturated fatty acids, dietary fiber, and protein have a significant impact on the composition of bile salts and short-chain fatty acids through catabolic processes, thereby influencing the gut microbiota. Additionally, these compounds also have an effect on the ratio of Firmicutes to Bacteroides, as well as the abundance of specific species like Akkermansia muciniphila. The gut microbiota has been found to play a role in altering the absorption and metabolism of nutrients, bioactive compounds, and drugs, adding another layer of complexity to the interaction between food and gut microbiota, which often requires long-term adaptation to yield substantial outcomes. In conclusion, understanding the relationship between food compounds and gut microbiota can offer valuable insights into the potential therapeutic applications of food and dietary interventions in various diseases and health conditions.
Collapse
Affiliation(s)
- Yijun Zheng
- Clinical Pharmacy (Sino-Foreign Cooperation) Class, School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Chunyin Qin
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Mingchun Wen
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Liang Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei 230036, China; (C.Q.); (M.W.)
| | - Weinan Wang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Blvd, Dongguan 523808, China
| |
Collapse
|
95
|
Alagiakrishnan K, Morgadinho J, Halverson T. Approach to the diagnosis and management of dysbiosis. Front Nutr 2024; 11:1330903. [PMID: 38706561 PMCID: PMC11069313 DOI: 10.3389/fnut.2024.1330903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/12/2024] [Indexed: 05/07/2024] Open
Abstract
All microorganisms like bacteria, viruses and fungi that reside within a host environment are considered a microbiome. The number of bacteria almost equal that of human cells, however, the genome of these bacteria may be almost 100 times larger than the human genome. Every aspect of the physiology and health can be influenced by the microbiome living in various parts of our body. Any imbalance in the microbiome composition or function is seen as dysbiosis. Different types of dysbiosis are seen and the corresponding symptoms depend on the site of microbial imbalance. The contribution of the intestinal and extra-intestinal microbiota to influence systemic activities is through interplay between different axes. Whole body dysbiosis is a complex process involving gut microbiome and non-gut related microbiome. It is still at the stage of infancy and has not yet been fully understood. Dysbiosis can be influenced by genetic factors, lifestyle habits, diet including ultra-processed foods and food additives, as well as medications. Dysbiosis has been associated with many systemic diseases and cannot be diagnosed through standard blood tests or investigations. Microbiota derived metabolites can be analyzed and can be useful in the management of dysbiosis. Whole body dysbiosis can be addressed by altering lifestyle factors, proper diet and microbial modulation. The effect of these interventions in humans depends on the beneficial microbiome alteration mostly based on animal studies with evolving evidence from human studies. There is tremendous potential for the human microbiome in the diagnosis, treatment, and prognosis of diseases, as well as, for the monitoring of health and disease in humans. Whole body system-based approach to the diagnosis of dysbiosis is better than a pure taxonomic approach. Whole body dysbiosis could be a new therapeutic target in the management of various health conditions.
Collapse
Affiliation(s)
| | - Joao Morgadinho
- Kaye Edmonton Clinic, Alberta Health Services, Edmonton, AB, Canada
| | - Tyler Halverson
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
96
|
Baghel K, Khan A, Kango N. Role of Synbiotics (Prebiotics and Probiotics) as Dietary Supplements in Type 2 Diabetes Mellitus Induced Health Complications. J Diet Suppl 2024; 21:677-708. [PMID: 38622882 DOI: 10.1080/19390211.2024.2340509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Diabetes is a metabolic disorder whose prevalence has become a worrying condition in recent decades. Chronic diabetes can result in serious health conditions such as impaired kidney function, stroke, blindness, and myocardial infarction. Despite a variety of currently available treatments, cases of diabetes and its complications are on the rise. This review article provides a comprehensive account of the ameliorative effect of prebiotics and probiotics individually or in combination i.e. synbiotics on health complications induced by Type 2 Diabetes Mellitus (T2DM). Recent advances in the field underscore encouraging outcomes suggesting the consumption of synbiotics leads to favorable changes in the gut microbiota. These changes result in the production of bioactive metabolites such as short-chain fatty acids (crucial for lowering blood sugar levels), reducing inflammation, preventing insulin resistance, and encouraging the release of glucagon-like peptide-1 in the host. Notably, novel strategies supplementing synbiotics to support gut microbiota are gaining attraction as pivotal interventions in mitigating T2DM-induced health complications. Thus, by nurturing a symbiotic relationship between prebiotics and probiotics i.e. synbiotics, these interventions hold promise in reshaping the microbial landscape of the gut thereby offering a multifaceted approach to managing T2DM and its associated morbidities. Supporting the potential of synbiotics underscores a paradigm shift toward holistic and targeted interventions in diabetes management, offering prospects for improved outcomes and enhanced quality of life for affected individuals. Nevertheless, more research needs to be done to better understand the single and multispecies pre/pro and synbiotics in the prevention and management of T2DM-induced health complications.
Collapse
Affiliation(s)
- Kalpana Baghel
- Department of Microbiology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
- Department of Zoology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| | - Aamir Khan
- Department of Zoology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| | - Naveen Kango
- Department of Microbiology, School of Biological Sciences, Dr Harisingh Gour Vishwavidyalaya (A Central University), Sagar, MP, India
| |
Collapse
|
97
|
Iddrisu I, Monteagudo-Mera A, Poveda C, Shahzad M, Walton GE, Andrews SC. A review of the effect of iron supplementation on the gut microbiota of children in developing countries and the impact of prebiotics. Nutr Res Rev 2024:1-9. [PMID: 38586996 DOI: 10.1017/s0954422424000118] [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: 04/09/2024]
Abstract
Iron is essential for many physiological functions of the body, and it is required for normal growth and development. Iron deficiency (ID) is the most common form of micronutrient malnutrition and is particularly prevalent in infants and young children in developing countries. Iron supplementation is considered the most effective strategy to combat the risk of ID and ID anaemia (IDA) in infants, although iron supplements cause a range of deleterious gut-related problems in malnourished children. The purpose of this review is to assess the available evidence on the effect of iron supplementation on the gut microbiota during childhood ID and to further assess whether prebiotics offer any benefits for iron supplementation. Prebiotics are well known to improve gut-microbial health in children, and recent reports indicate that prebiotics can mitigate the adverse gut-related effects of iron supplementation in children with ID and IDA. Thus, provision of prebiotics alongside iron supplements has the potential for an enhanced strategy for combatting ID and IDA among children in the developing world. However, further understanding is required before the benefit of such combined treatments of ID in nutritionally deprived children across populations can be fully confirmed. Such enhanced understanding is of high relevance in resource-poor countries where ID, poor sanitation and hygiene, alongside inadequate access to good drinking water and poor health systems, are serious public health concerns.
Collapse
Affiliation(s)
- Ishawu Iddrisu
- Rose Ward, Prospect Park Hospital, Berkshire Healthcare NHS Foundation Trust, Reading, RG30 4EJ, UK
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| | - Andrea Monteagudo-Mera
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Carlos Poveda
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Muhammed Shahzad
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
- Faculty of Dentistry, Zarqa University, Zarqa, 13110, Jordan
| | - Gemma E Walton
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Simon C Andrews
- School of Biological Sciences, University of Reading, Whiteknights, Reading, RG6 6EX, UK
| |
Collapse
|
98
|
Verma A, Bhagchandani T, Rai A, Nikita, Sardarni UK, Bhavesh NS, Gulati S, Malik R, Tandon R. Short-Chain Fatty Acid (SCFA) as a Connecting Link between Microbiota and Gut-Lung Axis-A Potential Therapeutic Intervention to Improve Lung Health. ACS OMEGA 2024; 9:14648-14671. [PMID: 38585101 PMCID: PMC10993281 DOI: 10.1021/acsomega.3c05846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 04/09/2024]
Abstract
The microbiome is an integral part of the human gut, and it plays a crucial role in the development of the immune system and homeostasis. Apart from the gut microbiome, the airway microbial community also forms a distinct and crucial part of the human microbiota. Furthermore, several studies indicate the existence of communication between the gut microbiome and their metabolites with the lung airways, called "gut-lung axis". Perturbations in gut microbiota composition, termed dysbiosis, can have acute and chronic effects on the pathophysiology of lung diseases. Microbes and their metabolites in lung stimulate various innate immune pathways, which modulate the expression of the inflammatory genes in pulmonary leukocytes. For instance, gut microbiota-derived metabolites such as short-chain fatty acids can suppress lung inflammation through the activation of G protein-coupled receptors (free fatty acid receptors) and can also inhibit histone deacetylase, which in turn influences the severity of acute and chronic respiratory diseases. Thus, modulation of the gut microbiome composition through probiotic/prebiotic usage and fecal microbiota transplantation can lead to alterations in lung homeostasis and immunity. The resulting manipulation of immune cells function through microbiota and their key metabolites paves the way for the development of novel therapeutic strategies in improving the lung health of individuals affected with various lung diseases including SARS-CoV-2. This review will shed light upon the mechanistic aspect of immune system programming through gut and lung microbiota and exploration of the relationship between gut-lung microbiome and also highlight the therapeutic potential of gut microbiota-derived metabolites in the management of respiratory diseases.
Collapse
Affiliation(s)
- Anjali Verma
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Tannu Bhagchandani
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ankita Rai
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nikita
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Urvinder Kaur Sardarni
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Neel Sarovar Bhavesh
- Transcription
Regulation Group, International Centre for
Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Sameer Gulati
- Department
of Medicine, Lady Hardinge Medical College
(LHMC), New Delhi 110058, India
| | - Rupali Malik
- Department
of Medicine, Vardhman Mahavir Medical College
and Safdarjung Hospital, New Delhi 110029, India
| | - Ravi Tandon
- Laboratory
of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
99
|
Revankar NA, Negi PS. Biotics: An emerging food supplement for health improvement in the era of immune modulation. Nutr Clin Pract 2024; 39:311-329. [PMID: 37466413 DOI: 10.1002/ncp.11036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/27/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
The involvement of the commensal microbiota in immune function is a multifold process. Biotics, such as probiotics, prebiotics, synbiotics, and paraprobiotics, have been subjected to animal and human trials demonstrating the association between gut microbes and immunity biomarkers leading to improvement in overall health. In recent years, studies on human microbiome interaction have established the multifarious role of biotics in maintaining overall health. The consumption of biotics has been extensively reported to help in maintaining microbial diversity, enhancing gut-associated mucosal immune homeostasis, and providing protection against a wide range of lifestyle disorders. However, the establishment of biotics as an alternative therapy for a range of health conditions is yet to be ascertained. Despite the fact that scientific literature has demonstrated the correlation between biotics and immune modulation, most in vivo and in vitro reports are inconclusive on the dosage required. This review provides valuable insights into the immunomodulatory effects of biotics consumption based on evidence obtained from animal models and clinical trials. Furthermore, we highlight the optimal dosages of biotics that have been reported to deliver maximum health benefits. By identifying critical research gaps, we have suggested a roadmap for future investigations to advance our understanding of the intricate crosstalk between biotics and immune homeostasis.
Collapse
Affiliation(s)
- Neelam A Revankar
- Department of Fruit and Vegetables Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pradeep S Negi
- Department of Fruit and Vegetables Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
100
|
Zeng Z, Tang W. Gut microbiota: A potential player in psychiatric symptoms during COVID-19. World J Biol Psychiatry 2024; 25:267-280. [PMID: 38607962 DOI: 10.1080/15622975.2024.2342846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES This study aims to explore the potential interconnections among gut microbiota, COVID-19 infection, depression and anxiety disorder. Additionally, it tries to assess potential therapeutic interventions that may improve the dysbiosis of gut microbiota. METHODS To achieve these objectives, we reviewed existing literature, encompassing studies and critical reviews that intersect the domains of gut microbiota, COVID-19, depression and anxiety disorders. RESULTS The findings highlight a notable correlation between the dysbiosis of gut microbiota and psychiatric symptoms in the context of COVID-19. Specifically, there is a marked reduction in the populations of bacteria that generate anti-inflammatory short-chain fatty acids (SCFAs), alongside a rise in the prevalence of gut bacterial clusters linked to inflammatory processes. Furthermore, several potential treatment strategies were summarised for improving the dysbiosis. CONCLUSIONS Gut microbiota plays a significant role in psychiatric symptoms during COVID-19, which has significant implications for the study and prevention of psychiatric symptoms in major epidemic diseases.
Collapse
Affiliation(s)
- Zijie Zeng
- Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | | |
Collapse
|