1
|
Slater AS, Hickey RM, Davey GP. Interactions of human milk oligosaccharides with the immune system. Front Immunol 2025; 15:1523829. [PMID: 39877362 PMCID: PMC11772441 DOI: 10.3389/fimmu.2024.1523829] [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: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Human milk oligosaccharides (HMOs) are abundant, diverse and complex sugars present in human breast milk. HMOs are well-characterized barriers to microbial infection and by modulating the human microbiome they are also thought to be nutritionally beneficial to the infant. The structural variety of over 200 HMOs, including neutral, fucosylated and sialylated forms, allows them to interact with the immune system in various ways. Clinically, HMOs impact allergic diseases, reducing autoimmune and inflammatory responses, and offer beneficial support to the preterm infant immune health. This review examines the HMO composition and associated immunomodulatory effects, including interactions with immune cell receptors and gut-associated immune responses. These immunomodulatory properties highlight the potential for HMO use in early stage immune development and for use as novel immunotherapeutics. HMO research is rapidly evolving and promises innovative treatments for immune-related conditions and improved health outcomes.
Collapse
Affiliation(s)
- Alanna S. Slater
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Gavin P. Davey
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
van de Worp WRPH, Theys J, Wolfs CJA, Verhaegen F, Schols AMWJ, van Helvoort A, Langen RCJ. Targeted nutritional intervention attenuates experimental lung cancer cachexia. J Cachexia Sarcopenia Muscle 2024; 15:1664-1676. [PMID: 38965830 PMCID: PMC11446694 DOI: 10.1002/jcsm.13520] [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: 08/25/2023] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Cachexia, a syndrome with high prevalence in non-small cell lung cancer patients, impairs quality of life and reduces tolerance and responsiveness to cancer therapy resulting in decreased survival. Optimal nutritional care is pivotal in the treatment of cachexia and a recommended cornerstone of multimodal therapy. Here, we investigated the therapeutic effect of an intervention diet consisting of a specific combination of high protein, leucine, fish oil, vitamin D, galacto-oligosaccharides, and fructo-oligosaccharides on the development and progression of cachexia in an orthotopic lung cancer mouse model. METHODS Eleven-week-old male 129S2/Sv mice were orthotopically implanted with 344P lung epithelial tumour cells or vehicle (control). Seven days post-implantation tumour-bearing (TB) mice were allocated to either intervention- or isocaloric control diet. Cachexia was defined as 5 days of consecutive body weight loss, after which mice were euthanized for tissue analyses. RESULTS TB mice developed cachexia accompanied by significant loss of skeletal muscle mass and epididymal fat mass compared with sham operated mice. The cachectic endpoint was significantly delayed (46.0 ± 15.2 vs. 34.7 ± 11.4 days), and the amount (-1.57 ± 0.62 vs. -2.13 ± 0.57 g) and progression (-0.26 ± 0.14 vs. -0.39 ± 0.11 g/day) of body weight loss were significantly reduced by the intervention compared with control diet. Moreover, systemic inflammation (pentraxin-2 plasma levels) and alterations in molecular markers for proteolysis and protein synthesis, indicative of muscle atrophy signalling in TB-mice, were suppressed in skeletal muscle by the intervention diet. CONCLUSIONS Together, these data demonstrate the potential of this multinutrient intervention, targeting multiple components of cachexia, as integral part of lung cancer management.
Collapse
Affiliation(s)
- Wouter R. P. H. van de Worp
- Department of Respiratory Medicine, NUTRIM – Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Jan Theys
- Department of Precision Medicine, GROW – Institute for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Cecile J. A. Wolfs
- Department of radiation Oncology (Maastro), GROW – Institute for Oncology and ReproductionMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Frank Verhaegen
- Department of radiation Oncology (Maastro), GROW – Institute for Oncology and ReproductionMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Annemie M. W. J. Schols
- Department of Respiratory Medicine, NUTRIM – Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Ardy van Helvoort
- Department of Respiratory Medicine, NUTRIM – Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Danone Nutricia ResearchUtrechtThe Netherlands
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM – Institute of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
3
|
Zhang W, Mackay CR, Gershwin ME. Immunomodulatory Effects of Microbiota-Derived Short-Chain Fatty Acids in Autoimmune Liver Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1629-1639. [PMID: 37186939 PMCID: PMC10188201 DOI: 10.4049/jimmunol.2300016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/01/2023] [Indexed: 05/17/2023]
Abstract
Nonpathogenic commensal microbiota and their metabolites and components are essential to maintain a tolerogenic environment and promote beneficial health effects. The metabolic environment critically impacts the outcome of immune responses and likely impacts autoimmune and allergic responses. Short-chain fatty acids (SCFAs) are the main metabolites produced by microbial fermentation in the gut. Given the high concentration of SCFAs in the gut and portal vein and their broad immune regulatory functions, SCFAs significantly influence immune tolerance and gut-liver immunity. Alterations of SCFA-producing bacteria and SCFAs have been identified in a multitude of inflammatory diseases. These data have particular significance in primary biliary cholangitis, primary sclerosing cholangitis, and autoimmune hepatitis because of the close proximity of the liver to the gut. In this focused review, we provide an update on the immunologic consequences of SCFA-producing microbiota and in particular on three dominant SCFAs in autoimmune liver diseases.
Collapse
Affiliation(s)
- Weici Zhang
- Division of Rheumatology, Allergy, and Clinical Immunology, School of Medicine, University of California Davis, CA, USA
| | - Charles R. Mackay
- Department of Microbiology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, School of Medicine, University of California Davis, CA, USA
| |
Collapse
|
4
|
Luo S, Wang Y, Kang X, Liu P, Wang G. Research progress on the association between mastitis and gastrointestinal microbes in dairy cows and the effect of probiotics. Microb Pathog 2022; 173:105809. [PMID: 36183956 DOI: 10.1016/j.micpath.2022.105809] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 11/15/2022]
Abstract
Mastitis in dairy cows affects milk quality and thereby constrains the development of the dairy industry. A clear understanding of the pathogenesis of mastitis can help its treatment. Mastitis is caused by the invasion of pathogenic bacteria into the mammary gland through the mammary ducts. However, recent studies suggested that an endogenous entero-mammary pathway in dairy cattle might also be playing an important role in regulating mastitis. Also, probiotic intervention regulating host gut microbes has become an interesting tool to control mastitis. This review discusses the association of gastrointestinal microbes with mastitis and the mechanism of action of probiotics in dairy cows to provide new ideas for the management of mastitis in large-scale dairy farms.
Collapse
Affiliation(s)
- Shuangyan Luo
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Yuxia Wang
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Xinyun Kang
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Panpan Liu
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Guiqin Wang
- School of Agriculture, Ningxia University, 750021, Yinchuan, China.
| |
Collapse
|
5
|
Lampousi AM, Carlsson S, Löfvenborg JE. Dietary factors and risk of islet autoimmunity and type 1 diabetes: a systematic review and meta-analysis. EBioMedicine 2021; 72:103633. [PMID: 34656932 PMCID: PMC8523874 DOI: 10.1016/j.ebiom.2021.103633] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background Numerous dietary components have been linked to the development of islet autoimmunity (IA) and type 1 diabetes (T1D); however, no associations are firmly established. This systematic review and meta-analysis aimed to synthesize current knowledge on diet and incidence of IA and T1D. Methods Literature search was performed in Medline, Embase, and Cochrane Library, from inception until October 2020. Eligible studies had IA or T1D as outcome; any dietary exposure; case-control, cohort, or randomized controlled trial design; and hazard, risk, or odds ratios as measures of association. Summary relative risks (RR) and 95% confidence intervals (CI) were estimated with random-effects models. Certainty of evidence was assessed with GRADE. PROSPERO registration number: CRD42020212505. Findings Among 5935 identified records, 96 were eligible, and pooled estimates could be produced for 26 dietary factors. Evidence with moderate/high certainty indicated lower risk of T1D in relation to longer (≥6-12 vs <6-12 months, RR: 0⋅39, CI: 0⋅26-0⋅58, I2=43%) and exclusive (≥2-3 vs <2-3 months, RR: 0⋅68, CI: 0⋅58-0⋅80, I2=0%) breastfeeding, later introduction to gluten (3-6 vs <3-5 months, RR: 0⋅36, CI: 0⋅17-0⋅75, I2=0%), cow's milk (≥2-3 vs <2-3 months, RR: 0⋅69, CI: 0⋅59-0⋅81, I2=0%), and fruit (4-6 vs <4-5 months, RR: 0⋅47, CI: 0⋅25-0⋅86, I2=0%). Higher childhood intake of cow's milk was associated with increased risk of both IA (per 2-3 portions/day, RR: 1⋅25, CI: 1⋅06-1⋅47, I2=0%) and T1D (≥2-3 vs <2-3 glasses/day, RR: 1⋅81, CI: 1⋅12-2⋅91, I2=31%). For the remaining dietary factors investigated, there was no association, or the evidence was of low certainty. Interpretation This study suggests that breastfeeding and late introduction of gluten, fruit, and cow's milk may reduce the risk of T1D, whereas high childhood cow's milk intake may increase it. Funding Swedish Research Council, Swedish Research Council for Health, Working Life and Welfare (FORTE), Novo Nordisk Foundation, and Swedish Diabetes Foundation.
Collapse
Affiliation(s)
- Anna-Maria Lampousi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
6
|
Jia LL, Zhang M, Liu H, Sun J, Pan LL. Early-life fingolimod treatment improves intestinal homeostasis and pancreatic immune tolerance in non-obese diabetic mice. Acta Pharmacol Sin 2021; 42:1620-1629. [PMID: 33473182 PMCID: PMC8463616 DOI: 10.1038/s41401-020-00590-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
Fingolimod has beneficial effects on multiple diseases, including type 1 diabetes (T1D) and numerous preclinical models of colitis. Intestinal dysbiosis and intestinal immune dysfunction contribute to disease pathogenesis of T1D. Thus, the beneficial effect of fingolimod on T1D may occur via the maintenance of intestinal homeostasis to some extent. Herein, we investigated the role of fingolimod in intestinal dysfunction in non-obese diabetic (NOD) mice and possible mechanisms. NOD mice were treated with fingolimod (1 mg · kg-1 per day, i.g.) from weaning (3-week-old) to 31 weeks of age. We found that fingolimod administration significantly enhanced the gut barrier (evidenced by enhanced expression of tight junction proteins and reduced intestinal permeability), attenuated intestinal microbial dysbiosis (evidenced by the reduction of enteric pathogenic Proteobacteria clusters), as well as intestinal immune dysfunction (evidenced by inhibition of CD4+ cells activation, reduction of T helper type 1 cells and macrophages, and the expansion of regulatory T cells). We further revealed that fingolimod administration suppressed the activation of CD4+ cells and the differentiation of T helper type 1 cells, promoted the expansion of regulatory T cells in the pancreas, which might contribute to the maintenance of pancreatic immune tolerance and the reduction of T1D incidence. The protection might be due to fingolimod inhibiting the toll-like receptor 2/4/nuclear factor-κB/NOD-like receptor protein 3 inflammasome pathway in the colon. Collectively, early-life fingolimod treatment attenuates intestinal microbial dysbiosis and intestinal immune dysfunction in the T1D setting, which might contribute to its anti-diabetic effect.
Collapse
Affiliation(s)
- Ling-Ling Jia
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Ming Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - He Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
7
|
Sekerel BE, Bingol G, Cullu Cokugras F, Cokugras H, Kansu A, Ozen H, Tamay Z. An Expert Panel Statement on the Beneficial Effects of Human Milk Oligosaccharides (HMOs) in Early Life and Potential Utility of HMO-Supplemented Infant Formula in Cow's Milk Protein Allergy. J Asthma Allergy 2021; 14:1147-1164. [PMID: 34594114 PMCID: PMC8478436 DOI: 10.2147/jaa.s323734] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
This review by pediatric gastroenterology and allergy-immunology experts aimed to address the biological roles of human milk oligosaccharides (HMOs) and the potential utility of HMOs in prevention of allergy with particular emphasis on cow's milk protein allergy (CMPA). The participating experts consider HMOs amongst the most critical bioactive components of human milk, which act as antimicrobials and antivirals by preventing pathogen adhesion to epithelial cells, as intestinal epithelial cell modulators by enhancing maturation of intestinal mucosa and intestinal epithelial barrier function, as prebiotics by promoting healthy microbiota composition and as immunomodulators by modulating immune cells indirectly and directly. Accordingly, the participating experts consider the proposed link between HMOs and prevention of allergy to be primarily based on the impact of HMO on gut microbiota, intestinal mucosal barrier, immunomodulation and immune maturation. Along with the lower risk of respiratory and gastrointestinal infections, HMO-supplemented formulas seem to be promising alternatives in the management of CMPA. Nonetheless, the effects of individual as well as complex mixtures of HMO in terms of clear clinical and immunological effects and tolerance development need to be further explored to fully realize the immunomodulatory mechanisms and the potential for HMOs in prevention of allergic diseases and CMPA.
Collapse
Affiliation(s)
- Bulent Enis Sekerel
- Division of Pediatric Allergy, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Gulbin Bingol
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Fugen Cullu Cokugras
- Division of Pediatric Gastroenterology, Department of Pediatrics, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Haluk Cokugras
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Aydan Kansu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Hasan Ozen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Zeynep Tamay
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
8
|
Verduci E, Mameli C, Amatruda M, Petitti A, Vizzuso S, El Assadi F, Zuccotti G, Alabduljabbar S, Terranegra A. Early Nutrition and Risk of Type 1 Diabetes: The Role of Gut Microbiota. Front Nutr 2021; 7:612377. [PMID: 33425976 PMCID: PMC7785819 DOI: 10.3389/fnut.2020.612377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) appears most frequently in childhood, with an alarming increasing incidence in the last decades. Although the genetic predisposition is a major risk factor, it cannot solely explain the complex etiology of T1D which is still not fully understood. In this paper, we reviewed the most recent findings on the role of early nutrition and the involvement of the gut microbiota in the etiopathogenesis of T1D. The main conclusions that are withdrawn from the current literature regarding alleviating the risk of developing T1D through nutrition are the encouragement of long-term breast-feeding for at least the first 6 months of life and the avoidance of early complementary foods and gluten introduction (before 4 months of age) as well as cow milk introduction before 12 months of life. These detrimental feeding habits create a gut microbiota dysbiotic state that can contribute to the onset of T1D in infancy. Finally, we discussed the possibility to introduce probiotics, prebiotics and post-biotics in the prevention of T1D.
Collapse
Affiliation(s)
- Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Chiara Mameli
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Matilde Amatruda
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Agnese Petitti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | - Sara Vizzuso
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Farah El Assadi
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Milan, Italy
| | | | | |
Collapse
|
9
|
van de Worp WRPH, Schols AMWJ, Theys J, van Helvoort A, Langen RCJ. Nutritional Interventions in Cancer Cachexia: Evidence and Perspectives From Experimental Models. Front Nutr 2020; 7:601329. [PMID: 33415123 PMCID: PMC7783418 DOI: 10.3389/fnut.2020.601329] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer cachexia is a complex metabolic syndrome characterized by involuntary skeletal muscle loss and is associated with poor clinical outcome, decreased survival and negatively influences cancer therapy. No curative treatments are available for cancer cachexia, but nutritional intervention is recommended as a cornerstone of multimodal therapy. Optimal nutritional care is pivotal in the treatment of cancer cachexia, and the effects of nutrients may extend beyond provision of adequate energy uptake, targeting different mechanisms or metabolic pathways that are affected or deregulated by cachexia. The evidence to support this notion derived from nutritional intervention studies in experimental models of cancer cachexia is systematically discussed in this review. Moreover, experimental variables and readout parameters to determine skeletal muscle wasting and cachexia are methodologically evaluated to allow critical comparison of similar studies. Single- and multinutrient intervention studies including qualitative modulation of dietary protein, dietary fat, and supplementation with specific nutrients, such as carnitine and creatine, were reviewed for their efficacy to counteract muscle mass loss and its underlying mechanisms in experimental cancer cachexia. Numerous studies showed favorable effects on impaired protein turnover and related metabolic abnormalities of nutritional supplementation in parallel with a beneficial impact on cancer-induced muscle wasting. The combination of high quality nutrients in a multitargeted, multinutrient approach appears specifically promising, preferentially as a multimodal intervention, although more studies investigating the optimal quantity and combination of nutrients are needed. During the review process, a wide variation in timing, duration, dosing, and route of supplementation, as well as a wide variation in animal models were observed. Better standardization in dietary design, and the development of experimental models that better recapitulate the etiology of human cachexia, will further facilitate successful translation of experimentally-based multinutrient, multimodal interventions into clinical practice.
Collapse
Affiliation(s)
- Wouter R P H van de Worp
- Department of Respiratory Medicine, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Jan Theys
- Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ardy van Helvoort
- Department of Respiratory Medicine, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands.,Danone Nutricia Research, Utrecht, Netherlands
| | - Ramon C J Langen
- Department of Respiratory Medicine, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
10
|
Raspini B, Porri D, De Giuseppe R, Chieppa M, Liso M, Cerbo RM, Civardi E, Garofoli F, Monti MC, Vacca M, De Angelis M, Cena H. Prenatal and postnatal determinants in shaping offspring's microbiome in the first 1000 days: study protocol and preliminary results at one month of life. Ital J Pediatr 2020; 46:45. [PMID: 32293504 PMCID: PMC7158098 DOI: 10.1186/s13052-020-0794-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fetal programming during in utero life defines the set point of physiological and metabolic responses that lead into adulthood; events happening in "the first 1,000 days" (from conception to 2-years of age), play a role in the development of non-communicable diseases (NCDs). The infant gut microbiome is a highly dynamic organ, which is sensitive to maternal and environmental factors and is one of the elements driving intergenerational NCDs' transmission. The A.MA.MI (Alimentazione MAmma e bambino nei primi MIlle giorni) project aims at investigating the correlation between several factors, from conception to the first year of life, and infant gut microbiome composition. We described the study design of the A.MA.MI study and presented some preliminary results. METHODS A.MA.MI is a longitudinal, prospective, observational study conducted on a group of mother-infant pairs (n = 60) attending the Neonatal Unit, Fondazione IRCCS Policlinico San Matteo, Pavia (Italy). The study was planned to provide data collected at T0, T1, T2 and T3, respectively before discharge, 1,6 and 12 months after birth. Maternal and infant anthropometric measurements were assessed at each time. Other variables evaluated were: pre-pregnancy/gestational weight status (T0), maternal dietary habits/physical activity (T1-T3); infant medical history, type of feeding, antibiotics/probiotics/supplements use, environment exposures (e.g cigarette smoking, pets, environmental temperature) (T1-T3). Infant stool samples were planned to be collected at each time and analyzed using metagenomics 16S ribosomal RNA gene sequence-based methods. RESULTS Birth mode (cesarean section vs. vaginal delivery) and maternal pre pregnancy BMI (BMI < 25 Kg/m2 vs. BMI ≥ 25 Kg/m2), significant differences were found at genera and species levels (T0). Concerning type of feeding (breastfed vs. formula-fed), gut microbiota composition differed significantly at genus and species level (T1). CONCLUSION These preliminary and explorative results confirmed that pre-pregnancy, mode of delivery and infant factors likely impact infant microbiota composition at different levels. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT04122612.
Collapse
Affiliation(s)
- Benedetta Raspini
- Dietetics and Clinical Nutrition Laboratory - Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, 27100, Pavia, Italy.
| | - Debora Porri
- Dietetics and Clinical Nutrition Laboratory - Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, 27100, Pavia, Italy
| | - Rachele De Giuseppe
- Dietetics and Clinical Nutrition Laboratory - Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, 27100, Pavia, Italy
| | - Marcello Chieppa
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Castellana, 70013, Grotte, BA, Italy.,European Biomedical Research Institute of Salerno EBRIS, 84125, Salerno, Italy
| | - Marina Liso
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Castellana, 70013, Grotte, BA, Italy
| | - Rosa Maria Cerbo
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Civardi
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesca Garofoli
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Cristina Monti
- Department of Public Health, Experimental and Forensic Medicine - Unit of Biostatistics and Clinical Epidemiology, University of Pavia, 27100, Pavia, Italy
| | - Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Hellas Cena
- Dietetics and Clinical Nutrition Laboratory - Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Bassi 21, 27100, Pavia, Italy.,Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
11
|
Yahaya T, Shemishere U. Association between Bioactive Molecules in Breast Milk and Type 1 Diabetes Mellitus. Sultan Qaboos Univ Med J 2020; 20:e5-e12. [PMID: 32190364 PMCID: PMC7065699 DOI: 10.18295/squmj.2020.20.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/23/2019] [Accepted: 11/03/2019] [Indexed: 02/05/2023] Open
Abstract
The association between breastfeeding and type 1 diabetes mellitus (T1DM) is controversial. However, several recent studies have established a link between these two factors, necessitating a need to review this subject to raise public awareness. Current research indicates that breast milk contains a variety of bioactive substances including immunoglobulins, oligosaccharides, insulin, lactoferrin, lysozyme, cytokines, epidermal growth factors, leukocytes, nucleotides, beneficial bacteria and vitamins. Such substances strengthen the breastfeeding infant's immune system, both directly, by increasing gut microbiota diversity and attacking harmful bacteria and pro-inflammatory molecules, and indirectly, by increasing thymus performance. Accordingly, a lack of or inadequate breastfeeding may predispose infants to several autoimmune disorders, including T1DM. Nursing mothers and caregivers are therefore advised to follow optimal breastfeeding practices prior to introducing complementary foods.
Collapse
Affiliation(s)
- Tajudeen Yahaya
- Department of Biology, Federal University Birnin Kebbi, Birnin Kebbi, Nigeria
| | - Ufuoma Shemishere
- Department of Biochemistry & Molecular Biology, Federal University Birnin Kebbi, Birnin Kebbi, Nigeria
| |
Collapse
|
12
|
Kobayashi N, Takahashi D, Takano S, Kimura S, Hase K. The Roles of Peyer's Patches and Microfold Cells in the Gut Immune System: Relevance to Autoimmune Diseases. Front Immunol 2019; 10:2345. [PMID: 31649668 PMCID: PMC6794464 DOI: 10.3389/fimmu.2019.02345] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023] Open
Abstract
Microfold (M) cells are located in the epithelium covering mucosa-associated lymphoid tissues, such as the Peyer's patches (PPs) of the small intestine. M cells actively transport luminal antigens to the underlying lymphoid follicles to initiate an immune response. The molecular machinery of M-cell differentiation and function has been vigorously investigated over the last decade. Studies have shed light on the role of M cells in the mucosal immune system and have revealed that antigen uptake by M cells contributes to not only mucosal but also systemic immune responses. However, M-cell studies usually focus on infectious diseases; the contribution of M cells to autoimmune diseases has remained largely unexplored. Accumulating evidence suggests that dysbiosis of the intestinal microbiota is implicated in multiple systemic diseases, including autoimmune diseases. This implies that the uptake of microorganisms by M cells in PPs may play a role in the pathogenesis of autoimmune diseases. We provide an outline of the current understanding of M-cell biology and subsequently discuss the potential contribution of M cells and PPs to the induction of systemic autoimmunity, beyond the mucosal immune response.
Collapse
Affiliation(s)
- Nobuhide Kobayashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan.,Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Shunsuke Takano
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Shunsuke Kimura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
13
|
Kumbhare SV, Patangia DVV, Patil RH, Shouche YS, Patil NP. Factors influencing the gut microbiome in children: from infancy to childhood. J Biosci 2019; 44:49. [PMID: 31180062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The human microbiota plays a crucial role in educating the immune system and influencing host health right since birth. Various maternal factors along with the vertical microbial transfer from the mother, as well as the horizontal environmental transmission and internal factors relating to the infant, play a crucial role in modulating the gut microbiota. The early life microflora is highly unstable and undergoes dynamic changes during the first few years, converging towards a more stabilized adult microbiota by co-evolving with the host by the age of 3-4 years. Microbiota studies have underlined the role of dysbiosis in developing several metabolic disorders like obesity, diabetes and immune-related disorders like asthma, to name a few. Thus, understanding early life microbial composition and various factors affecting the microbial community will provide a platform for developing strategies/techniques to maintain host health by restoring gut microbial flora. This review focuses on the factors that affect the microbial composition of the foetus in utero, during birth, infancy through childhood.
Collapse
Affiliation(s)
- Shreyas V Kumbhare
- National Centre for Cell Science, Savitribai Phule University of Pune Campus, Pune, Maharashtra 411007, India
| | | | | | | | | |
Collapse
|
14
|
|
15
|
Xiao L, van De Worp WR, Stassen R, van Maastrigt C, Kettelarij N, Stahl B, Blijenberg B, Overbeek SA, Folkerts G, Garssen J, Van't Land B. Human milk oligosaccharides promote immune tolerance via direct interactions with human dendritic cells. Eur J Immunol 2019; 49:1001-1014. [PMID: 30900752 PMCID: PMC6619030 DOI: 10.1002/eji.201847971] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/19/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Human milk oligosaccharides (HMOS) are a complex mixture of bioactive components supporting the immune development of breastfed‐infants. Dendritic cells (DCs) play a central role in the regulation of immune responses, being specialized in antigen presentation and driving T‐cell priming as well as differentiation. However, little is known about the direct effects of HMOS on human DC phenotypes and functions. Here, we report that HMOS mixture isolated from pooled human milk, induced semi‐maturation of human monocytes‐derived DCs (moDCs), and elevated levels of IL‐10, IL‐27 and IL‐6 but not IL‐12p70 and TNF‐α. Consistently, HMOS‐conditioned human moDCs promoted Treg generation from naïve CD4+ T cells. Interestingly, HMOS limited LPS‐induced maturation of human moDCs, while maintained IL‐10 and IL‐27 secretion and reduced LPS‐induced production of IL‐12p70, IL‐6 and TNF‐α. Furthermore, HMOS+LPS‐stimulated DCs induced a higher frequency of Tregs and increased IL‐10 production, while a reduction in Tbet+Th1 frequency and IFN‐γ production was detected as compared to LPS‐DCs. The regulatory effects of HMOS seemed to be mediated by interactions of HMOS with receptors, including but not limited to TLR4 and DC‐SIGN on human moDCs. In conclusion, HMOS contain tolerogenic factors influencing human moDCs and thereby modulating the development of the neonatal immune system.
Collapse
Affiliation(s)
- Ling Xiao
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Wouter Rph van De Worp
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.,Department of Respiratory Medicine, NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Roderick Stassen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Celine van Maastrigt
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Nienke Kettelarij
- Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands
| | - Bernd Stahl
- Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands
| | - Bernadet Blijenberg
- Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands
| | - Saskia A Overbeek
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.,Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands
| | - Gert Folkerts
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.,Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands
| | - Belinda Van't Land
- Departments of Immunology and of Human Milk Research & Analytical Science, Danone Nutricia Research, Utrecht, The Netherlands.,Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
16
|
Probiotics and Prebiotics for the Amelioration of Type 1 Diabetes: Present and Future Perspectives. Microorganisms 2019; 7:microorganisms7030067. [PMID: 30832381 PMCID: PMC6463158 DOI: 10.3390/microorganisms7030067] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 12/18/2022] Open
Abstract
Type 1-diabetes (T1D) is an autoimmune disease characterized by immune-mediated destruction of pancreatic beta (β)-cells. Genetic and environmental interactions play an important role in immune system malfunction by priming an aggressive adaptive immune response against β-cells. The microbes inhabiting the human intestine closely interact with the enteric mucosal immune system. Gut microbiota colonization and immune system maturation occur in parallel during early years of life; hence, perturbations in the gut microbiota can impair the functions of immune cells and vice-versa. Abnormal gut microbiota perturbations (dysbiosis) are often detected in T1D subjects, particularly those diagnosed as multiple-autoantibody-positive as a result of an aggressive and adverse immunoresponse. The pathogenesis of T1D involves activation of self-reactive T-cells, resulting in the destruction of β-cells by CD8⁺ T-lymphocytes. It is also becoming clear that gut microbes interact closely with T-cells. The amelioration of gut dysbiosis using specific probiotics and prebiotics has been found to be associated with decline in the autoimmune response (with diminished inflammation) and gut integrity (through increased expression of tight-junction proteins in the intestinal epithelium). This review discusses the potential interactions between gut microbiota and immune mechanisms that are involved in the progression of T1D and contemplates the potential effects and prospects of gut microbiota modulators, including probiotic and prebiotic interventions, in the amelioration of T1D pathology, in both human and animal models.
Collapse
|
17
|
Liang Y, Wang X, He D, You Q, Zhang T, Dong W, Fei J, Xing Y, Wu J. Ameliorating gut microenvironment through staphylococcal nuclease-mediated intestinal NETs degradation for prevention of type 1 diabetes in NOD mice. Life Sci 2019; 221:301-310. [PMID: 30776371 DOI: 10.1016/j.lfs.2019.02.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 12/20/2022]
Abstract
AIMS Recent studies have revealed that neutrophil extracellular traps (NETs) provide negative feedback in the progression to chronic inflammation and contribute to the pathogenesis of multiple autoimmune diseases including type 1 diabetes (T1D). In addition, accumulating evidences suggest that gut immunity play a key role in T1D pathogenesis. Our study aimed to evaluate whether staphylococcal nuclease (SNase) targeting intestinal NETs can ameliorate the intestinal inflammatory environment and protect against T1D development in non-obese diabetic(NOD) mice. MAIN METHODS Degradation of NETs with SNase in vitro was examined using SYTOX green assay. NOD/LtJ mice were oral administration of Lactococcus lactisl (L. lactis) pCYT: SNase and blood glucose levels were monitored weekly. Several biomarkers of NETs formation, gut leakage and inflammation were determined using a commercial ELISA kit. T Cell phenotypes in peripheral immune system were analyzed in flow cytometry and fecal samples were isolated to investigate intestinal microbiota. KEY FINDINGS The oral delivery of SNase by L. lactis can decrease the NETs levels and ameliorate inflammation both in the intestine and pancreatic islets and finally effectively regulate the blood glucose levels of NOD mice. Meanwhile, zonulin and lipopolysaccharide levels also reduced in SNase-fed NOD mice, suggesting SNase could improve gut barrier function via intestinal NETs degradation. Furthermore, the abundances of the intestinal microbiota and butyrate-producing gut bacteria were also increased with SNase treatment. SIGNIFICANCE SNase shows potential for intestinal NETs to prevent T1D based on the gut-pancreas axis.
Collapse
Affiliation(s)
| | | | | | - Qi You
- China Pharmaceutical University, China
| | | | | | | | - Yun Xing
- China Pharmaceutical University, China.
| | - Jie Wu
- China Pharmaceutical University, China.
| |
Collapse
|
18
|
Azagra-Boronat I, Massot-Cladera M, Knipping K, Van't Land B, Stahl B, Garssen J, Rodríguez-Lagunas MJ, Franch À, Castell M, Pérez-Cano FJ. Supplementation With 2'-FL and scGOS/lcFOS Ameliorates Rotavirus-Induced Diarrhea in Suckling Rats. Front Cell Infect Microbiol 2018; 8:372. [PMID: 30406046 PMCID: PMC6205980 DOI: 10.3389/fcimb.2018.00372] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
Rotavirus (RV) is considered to be the most common cause of gastroenteritis among infants aged less than 5 years old. Human milk bioactive compounds have the ability to modulate the diarrheic process caused by several intestinal pathogens. This study aimed to evaluate the potential protective role of a specific human milk oligosaccharide, 2′-fucosyllactose (2′-FL), a mixture of the prebiotic short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides 9:1 (GOS/FOS) and their combination (2′-FL+GOS/FOS) on RV-induced diarrhea in suckling rats. The nutritional intervention was performed from the second to the sixteenth day of life by oral gavage and on day 5 an RV strain was orally administered to induce infection. Fecal samples were scored daily to assess the clinical pattern of severity, incidence and duration of diarrhea. Blood and tissues were obtained at day 8 and 16 in order to evaluate the effects on the epithelial barrier and the mucosal and systemic immune responses. In the assessment of severity, incidence and duration of diarrhea, both 2′-FL and GOS/FOS displayed a beneficial effect in terms of amelioration. However, the mechanisms involved seemed to differ: 2′-FL displayed a direct ability to promote intestinal maturation and to enhance neonatal immune responses, while GOS/FOS induced an intestinal trophic effect and an RV-blocking action. The combination of 2′-FL and GOS/FOS showed additive effects in some variables. Therefore, it could be a good strategy to add these compounds in combination to infant formulas, to protect against human RV-induced diarrhea in children.
Collapse
Affiliation(s)
- Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| | - Karen Knipping
- Nutricia Research, Utrecht, Netherlands.,Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Belinda Van't Land
- Nutricia Research, Utrecht, Netherlands.,Department of Pediatric Immunology, University Medical Centre Utrecht/Wilhelmina Children's Hospital, Utrecht, Netherlands
| | | | - Johan Garssen
- Nutricia Research, Utrecht, Netherlands.,Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Maria José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| | - Francisco J Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), Barcelona, Spain
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW The incidence of type 1 diabetes (T1D) is rising drastically for the past decades at a rate that cannot be explained by genetic changes alone. Environmental changes are considered to be the main drivers of this change. Recently, the gut microbiota has been suggested as a missing link between known environmental disease modulators and T1D promotion. Lifestyle factors have changed over time and have altered the gut microbiota-host interaction affecting T1D development. The purpose of this review is to discuss recent data emphasizing the modulatory potential of early lifestyle factors on gut microbiota and to elucidate their implication for T1D. RECENT FINDINGS Recent findings show that lifestyle factors, especially those that affect the early establishment of gut homeostasis and the education of the immune system, are crucial disease modulators. Changing lifestyle factors affecting the early establishment of gut homeostasis are suggested to be key drivers of the rising T1D incidence.
Collapse
Affiliation(s)
- Elke Gülden
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
20
|
Zununi Vahed S, Moghaddas Sani H, Rahbar Saadat Y, Barzegari A, Omidi Y. Type 1 diabetes: Through the lens of human genome and metagenome interplay. Biomed Pharmacother 2018; 104:332-342. [PMID: 29775902 DOI: 10.1016/j.biopha.2018.05.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a genetic- and epigenetic-related disease from which a large population worldwide suffers. Some genetic factors along with various mutations related to the immune system for disease mechanism(s) have contrastively been determined. However, sometimes mechanisms have not been fully managed for the clarification of the initiation and/or progression of diseases to help patients. In the recent years, due to familiarity with the role of gut microbiota in the health, it has been found that the changes of the microbial balance in the industrialized societies can cause a battery of modern diseases, for which we have no specific definition of how they emerge. This work aims to explore the relationship between the human gut microbiota and the immune system along with their possible role in avoiding/emerging of type 1 diabetes (T1D) accompanied with the relation between genome and metagenome and their imbalance in causing T1D. Moreover, it provides novel view on how to balance the intestinal microbiota by lifestyle to hinder the mechanisms leading to T1D.
Collapse
Affiliation(s)
| | | | - Yalda Rahbar Saadat
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Xiao L, Leusink-Muis T, Kettelarij N, van Ark I, Blijenberg B, Hesen NA, Stahl B, Overbeek SA, Garssen J, Folkerts G, Van't Land B. Human Milk Oligosaccharide 2'-Fucosyllactose Improves Innate and Adaptive Immunity in an Influenza-Specific Murine Vaccination Model. Front Immunol 2018; 9:452. [PMID: 29593719 PMCID: PMC5854647 DOI: 10.3389/fimmu.2018.00452] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Background Human milk is uniquely suited to provide optimal nutrition and immune protection to infants. Human milk oligosaccharides are structural complex and diverse consisting of short chain and long chain oligosaccharides typically present in a 9:1 ratio. 2′-Fucosyllactose (2′FL) is one of the most prominent short chain oligosaccharides and is associated with anti-infective capacity of human milk. Aim To determine the effect of 2′FL on vaccination responsiveness (both innate and adaptive) in a murine influenza vaccination model and elucidate mechanisms involved. Methods A dose range of 0.25–5% (w/w) dietary 2′FL was provided to 6-week-old female C57Bl/6JOlaHsd mice 2 weeks prior primary and booster vaccination until the end of the experiment. Intradermal (i.d.) challenge was performed to measure the vaccine-specific delayed-type hypersensitivity (DTH). Antigen-specific antibody levels in serum as well as immune cell populations within several organs were evaluated using ELISA and flow cytometry, respectively. In an ex vivo restimulation assay, spleen cells were cocultured with influenza-loaded bone marrow-derived dendritic cells (BMDCs) to study the effects of 2′FL on vaccine-specific CD4+ and CD8+ T-cell proliferation and cytokine secretions. Furthermore, the direct immune regulatory effects of 2′FL were confirmed using in vitro BMDCs T-cell cocultures. Results Dietary 2′FL significantly (p < 0.05) enhanced vaccine specific DTH responses accompanied by increased serum levels of vaccine-specific immunoglobulin (Ig) G1 and IgG2a in a dose-dependent manner. Consistently, increased activation marker (CD27) expression on splenic B-cells was detected in mice receiving 2′FL as compared to control mice. Moreover, proliferation of vaccine-specific CD4+ and CD8+ T-cells, as well as interferon-γ production after ex vivo restimulation were significantly increased in spleen cells of mice receiving 2′FL as compared to control mice, which were in line with changes detected within dendritic cell populations. Finally, we confirmed a direct effect of 2′FL on the maturation status and antigen presenting capacity of BMDCs. Conclusion Dietary intervention with 2′FL improves both humoral and cellular immune responses to vaccination in mice, which might be attributed in part to the direct effects of 2′FL on immune cell differentiation.
Collapse
Affiliation(s)
- Ling Xiao
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Nienke Kettelarij
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands.,Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands
| | - Ingrid van Ark
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Bernadet Blijenberg
- Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands
| | - Nienke A Hesen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Bernd Stahl
- Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands
| | - Saskia A Overbeek
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands.,Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands
| | - Johan Garssen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands.,Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands
| | - Gert Folkerts
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Belinda Van't Land
- Department of Immunology, Human Milk Research, Nutricia Research, Utrecht, Netherlands.,Department of Paediatric Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
22
|
Xiao L, Van't Land B, Engen PA, Naqib A, Green SJ, Nato A, Leusink-Muis T, Garssen J, Keshavarzian A, Stahl B, Folkerts G. Human milk oligosaccharides protect against the development of autoimmune diabetes in NOD-mice. Sci Rep 2018; 8:3829. [PMID: 29497108 PMCID: PMC5832804 DOI: 10.1038/s41598-018-22052-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
Development of Type 1 diabetes (T1D) is influenced by non-genetic factors, such as optimal microbiome development during early life that "programs" the immune system. Exclusive and prolonged breastfeeding is an independent protective factor against the development of T1D, likely via bioactive components. Human Milk Oligosaccharides (HMOS) are microbiota modulators, known to regulate immune responses directly. Here we show that early life provision (only for a period of six weeks) of 1% authentic HMOS (consisting of both long-chain, as well as short-chain structures), delayed and suppressed T1D development in non-obese diabetic mice and reduced development of severe pancreatic insulitis in later life. These protective effects were associated with i) beneficial alterations in fecal microbiota composition, ii) anti-inflammatory microbiota-generating metabolite (i.e. short chain fatty acids (SCFAs)) changes in fecal, as well as cecum content, and iii) induction of anti-diabetogenic cytokine profiles. Moreover, in vitro HMOS combined with SCFAs induced development of tolerogenic dendritic cells (tDCs), priming of functional regulatory T cells, which support the protective effects detected in vivo. In conclusion, HMOS present in human milk are therefore thought to be vital in the protection of children at risk for T1D, supporting immune and gut microbiota development in early life.
Collapse
Affiliation(s)
- Ling Xiao
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Belinda Van't Land
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands.
- University Medical Center Utrecht, The Wilhelmina Children's Hospital, Laboratory of Translational Immunology, Utrecht, The Netherlands.
| | - Phillip A Engen
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
| | - Ankur Naqib
- DNA Services Facility, Research Resources Center, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Stefan J Green
- DNA Services Facility, Research Resources Center, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Angie Nato
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Thea Leusink-Muis
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Johan Garssen
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands
| | - Ali Keshavarzian
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
- Department of Pharmacology, Department of Physiology, Rush University Medical Center, Chicago, IL, USA
| | - Bernd Stahl
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands
| | - Gert Folkerts
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| |
Collapse
|
23
|
Triantis V, Bode L, van Neerven RJJ. Immunological Effects of Human Milk Oligosaccharides. Front Pediatr 2018; 6:190. [PMID: 30013961 PMCID: PMC6036705 DOI: 10.3389/fped.2018.00190] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Human milk oligosaccharides (HMOs) comprise a group of structurally complex, unconjugated glycans that are highly abundant in human milk. HMOs are minimally digested in the gastrointestinal tract and reach the colon intact, where they shape the microbiota. A small fraction of HMOs is absorbed, reaches the systemic circulation, and is excreted in urine. HMOs can bind to cell surface receptors expressed on epithelial cells and cells of the immune system and thus modulate neonatal immunity in the infant gut, and possibly also sites throughout the body. In addition, they have been shown to act as soluble decoy receptors to block the attachment of various microbial pathogens to cells. This review summarizes the current knowledge of the effects HMOs can have on infections, allergies, auto-immune diseases and inflammation, and will focus on the role of HMOs in altering immune responses through binding to immune-related receptors.
Collapse
Affiliation(s)
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Wageningen University and Research, Cell Biology and Immunology, Wageningen, Netherlands
| |
Collapse
|