1
|
Jia J, Bao P, Yu Q, Li N, Ren H, Chen Q, Yan P. Lactobacillus Re-Engineers Gut Microbiota to Overcome E. coli Colonization Resistance in Mice. Vet Sci 2025; 12:484. [PMID: 40431577 PMCID: PMC12115677 DOI: 10.3390/vetsci12050484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/28/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The intestinal health and functionality of animals play pivotal roles in nutrient digestion and absorption, as well as in maintaining defense against pathogenic invasions. These biological processes are modulated by various determinants, including husbandry conditions, dietary composition, and gut microbial ecology. The excessive use of anthropogenic antibiotics may disrupt intestinal microbiota composition, potentially leading to dysbiosis that directly compromises host homeostasis. While Lactobacillus species are recognized for their immunomodulatory properties, their precise mechanisms in regulating host anti-inflammatory gene expression and influencing mucosal layer maturation, particularly regarding E. coli colonization resistance, require further elucidation. To investigate the regulatory mechanisms of Lactobacillus in relation to intestinal architecture and function during E. coli infection, we established a colonic infection model using Bal b/c mice, conducting systematic analyses of intestinal morphology, inflammatory mediator profiles, and microbial community dynamics. Our results demonstrate that Lactobacillus supplementation (Pediococcus acidilactici) effectively mitigated E. coli O78-induced enteritis, with co-administration during infection facilitating the restoration of physiological parameters, including body mass, intestinal histoarchitecture, and microbial metabolic functions. Microbiome profiling revealed that the Lactobacillus intervention significantly elevated Lactococcus abundance while reducing Weissella populations (p < 0.05), concurrently enhancing metabolic pathways related to nutrient assimilation and environmental signal processing (including translation mechanisms, ribosomal biogenesis, amino acid transport metabolism, and energy transduction systems; p < 0.05). Mechanistically, Lactobacillus administration attenuated E. coli-induced intestinal pathology through multiple pathways: downregulating pro-inflammatory cytokine expression (IL-1β, IL-1α, and TNF-α), upregulating epithelial junctional complexes (Occludin, Claudin-1, and ZO-1), and stimulating mucin biosynthesis (MUC1 and MUC2; p < 0.05). These modifications collectively enhanced mucosal barrier integrity and promoted epithelial maturation. This investigation advances our comprehension of microbiota-host crosstalk during enteropathogenic infections under probiotic intervention, offering valuable insights for developing novel nutritional strategies and microbial management protocols in animal husbandry.
Collapse
Affiliation(s)
- Jianlei Jia
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.J.); (P.B.); (Q.Y.)
- School of Life Sciences, Qilu Normal University, Jinan 250200, China;
| | - Pengjia Bao
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.J.); (P.B.); (Q.Y.)
| | - Qinran Yu
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.J.); (P.B.); (Q.Y.)
| | - Ning Li
- Institute of Western Agriculture, The Chinese Academy of Agricultural Sciences, Changji 831100, China;
| | - Hao Ren
- Teamgene (Shandong) Agricultural Technology Co., Ltd., Zibo 255086, China;
| | - Qian Chen
- School of Life Sciences, Qilu Normal University, Jinan 250200, China;
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (J.J.); (P.B.); (Q.Y.)
| |
Collapse
|
2
|
Barros CA, Vieira TCRG. Lactoferrin as a Candidate Multifunctional Therapeutic in Synucleinopathies. Brain Sci 2025; 15:380. [PMID: 40309834 PMCID: PMC12025589 DOI: 10.3390/brainsci15040380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Lactoferrin (Lf) is a multifunctional glycoprotein with well-established antimicrobial, anti-inflammatory, and iron-binding properties. Emerging evidence suggests that Lf also plays a neuroprotective role, particularly in neurodegenerative disorders characterized by protein aggregation, such as Parkinson's disease (PD). Alpha-synuclein (aSyn) aggregation is a pathological hallmark of PD and other synucleinopathies, contributing to neuronal dysfunction and disease progression. Recent studies indicate that Lf may interfere with aSyn aggregation, iron chelation, and modulation of oxidative stress and neuroinflammation. Additionally, Lf's ability to cross the blood-brain barrier and its potential impact on the gut-brain axis highlight its promise as a therapeutic agent. This review explores Lf's mechanisms of action in synucleinopathies, its potential as a disease-modifying therapy, and innovative delivery strategies that could enhance its clinical applicability. By addressing the pathological and therapeutic dimensions of aSyn aggregation, we propose Lf as a compelling candidate for future research and clinical development in neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, RJ, Brazil;
| |
Collapse
|
3
|
Yu W, Wang W, Sheng X. Effect of A1 protein-free formula versus conventional formula on acute respiratory infections and diarrhea in toddlers: A Randomized Controlled Trial. J Pediatr Gastroenterol Nutr 2025; 80:705-713. [PMID: 39895612 PMCID: PMC11959105 DOI: 10.1002/jpn3.12473] [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: 09/16/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025]
Abstract
OBJECTIVES Acute respiratory infections (ARIs) and diarrhea are common in toddlers. Milk free of A1 β-casein (A1PF milk) may support the immune system, but few studies have investigated A1PF milk in toddler formula and any potential effects on ARI/diarrhea. This study's objective was to investigate the incidence of ARI and diarrhea with two toddler formulas, A1PF formula (A1PF) or conventional formula (CON), which differed in milk base and nutrient composition. METHODS This randomized, open-label, multicenter study (19 December 2022 to 17 May 2023) evaluated the occurrence of ARI and/or diarrhea in toddlers (aged 2-3 years) who consumed A1PF or CON over 90 days. RESULTS A total of 200 toddlers were enrolled, and 180 completed the study. The relative risk of ARI or diarrhea in the A1PF group versus the CON group did not differ significantly, but the median (interquartile range) ARI duration was significantly shorter in the A1PF group (3 [2-4] days vs. 5 [3-6] days, p = 0.012). At Day 90, toddlers consuming A1PF had significantly less severe bloating, gassiness, and fewer regurgitation events (all p < 0.05). Both formulas were well tolerated, and no serious adverse events were reported. CONCLUSION Toddlers who consumed A1PF had a reduced duration of ARI and improved diarrhea outcomes, reducing the burden on their families compared with toddlers who consumed CON. Although this is consistent with other studies, further research is required to determine whether these effects are solely attributable to the A1PF milk base or other differences between the formulas.
Collapse
Affiliation(s)
- Wen Yu
- Department of Child Health CareInternational Peace Maternal and Child Health Care HospitalShanghaiChina
| | - Weijie Wang
- Department of Child Health CareShanghai First Maternity and Infant HospitalShanghaiChina
| | - Xiaoyang Sheng
- Department of Developmental Behavioral Pediatric & Children Healthcare, Xinhua Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
4
|
Ding H, Qin J, Li Y, Dai L, Xu F, Liu Z, Shi X, Guan W, Sang J. Lactoferrin alleviates oxidative stress and endoplasmic reticulum stress induced by autoimmune thyroiditis by modulating the mTOR pathway in the thyroid. J Endocrinol Invest 2025; 48:861-876. [PMID: 39576556 DOI: 10.1007/s40618-024-02505-0] [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: 05/07/2024] [Accepted: 11/14/2024] [Indexed: 01/05/2025]
Abstract
Autoimmune thyroiditis (AITD) is a prevalent autoimmune disorder characterized by the immune system's attack on thyroid tissue, potentially leading to thyroid dysfunction, with a current lack of effective treatment modalities. Lactoferrin, a crucial functional dietary component obtainable from food sources, primarily exists in mammalian milk. We aim to investigate whether dietary supplementation with lactoferrin can protect the thyroid in Experimental Autoimmune Thyroiditis (EAT) rats. Our study reveals significantly elevated levels of oxidative stress (OS) and endoplasmic reticulum stress (ERS) in the AITD. Lactoferrin markedly reduces OS and infiltration of inflammatory cells in the thyroid tissue of EAT rats. Furthermore, lactoferrin inhibits ERS levels in the thyroid of EAT rats and alleviates cellular apoptosis. In vivo and in vitro experiments elucidate that its protective effect is primarily achieved through the inhibition of mTOR signaling pathway activation. In summary, lactoferrin, a nutrient readily obtainable from food sources, appears to be effective in mitigating thyroid damage in AITD.
Collapse
Affiliation(s)
- Haoran Ding
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Jiabo Qin
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yixuan Li
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Linghui Dai
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Fazhan Xu
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Zhijian Liu
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Xianbiao Shi
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China
| | - Wenxian Guan
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jianfeng Sang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
- Division of Thyroid Surgery, Department of General Surgery, The Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
5
|
Mokaram Doust Delkhah A. Blood transcriptomics identifies FEZ1 as a potential biomarker for inflammatory bowel disease. Comput Biol Med 2025; 187:109742. [PMID: 39894009 DOI: 10.1016/j.compbiomed.2025.109742] [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: 11/19/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION While the global burden of inflammatory bowel diseases (IBD) is increasing, the identification of novel therapeutic targets and biomarkers is of significant importance. In particular, blood transcriptomes provide a non-invasive source for biomarker discovery. Therefore, this study aimed to identify potential blood markers for IBD. METHODS By employing an integrated transcriptomics approach, four datasets obtained from blood specimens of patients with IBD were analyzed (GSE119600, GSE94648, GSE86434, and GSE71730). After determining differentially expressed genes (DEGs) in IBD, a protein-protein interaction (PPI) network was constructed, and regulatory miRNAs targeting hub genes were identified. Weighted gene co-expression network analysis (WGCNA) was carried out to determine IBD-specific modules. Subsequently, converging results from differential expression analysis and WGCNA were subjected to random forest (RF) decision tree-based and LASSO regression methods. Lastly, the diagnostic efficacy of genes highlighted by both machine learning methods was measured using receiver operating characteristic (ROC) analysis in the integrated dataset, in each individual dataset separately, and in external datasets (GSE276395, GSE169568, GSE112057, GSE100833, GSE33943, and GSE3365). RESULTS Downregulated TNF was identified as the central hub gene of the PPI network, and PRF1 was the only gene identified as a hub gene in a co-expressed gene module enriched in IBD. Following the identification of FEZ1 and NLRC5 among the top 10 genes by both RF and LASSO, ROC analysis demonstrated their acceptable diagnostic efficacy in the integrated data. However, only FEZ1 was considered a potential biomarker based on replication of the results in the external datasets. CONCLUSIONS The results of the present study suggest FEZ1 as a potential blood biomarker for IBD. While autophagy is currently the most convincing explanation for the involvement of FEZ1 in IBD, further investigations are required to elucidate its immunological role.
Collapse
|
6
|
Shafqat A, Li M, Zakirullah, Liu F, Tong Y, Fan J, Fan H. A comprehensive review of research advances in the study of lactoferrin to treat viral infections. Life Sci 2025; 361:123340. [PMID: 39730037 DOI: 10.1016/j.lfs.2024.123340] [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: 07/12/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Lactoferrin (Lf) is a naturally occurring glycoprotein known for its antiviral and antibacterial properties and is present in various physiological fluids. Numerous studies have demonstrated its antiviral effectiveness against multiple viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus (IFV), herpes simplex virus (HSV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Lf, a vital component of the mucosal defense system, plays a crucial role in inhibiting viral infection by binding to both host cells and viral particles, such as the Hepatitis C virus (HCV). This interaction enables Lf to keep viral particles away from their target cells, emphasizing its significance as a fundamental element of mucosal defense against viral infections. Additionally, Lf has the ability to modulate cytokine expression and enhance cellular immune responses. In the innate immune system, Lf serves as a unique iron transporter and helps suppress various pathogens like bacteria, fungi, and viruses. This article summarises the potential antiviral properties of Lf against various viruses, along with its other mentioned functions. The advancement of Lf-based therapies supports the homology of food and medicine, providing a promising avenue to address viral infections and other public health challenges.
Collapse
Affiliation(s)
- Amna Shafqat
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation, Guangzhou, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Junfen Fan
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin, China.
| |
Collapse
|
7
|
Giribaldi M, Nebbia S, Briard-Bion V, Jardin J, Ménard O, Dupont D, Coscia A, Cresi F, Lamberti C, Cavallarin L, Deglaire A. Peptidomic profile of human milk as influenced by fortification with different protein sources: An in vitro dynamic digestion simulation. Food Chem 2025; 462:140886. [PMID: 39213965 DOI: 10.1016/j.foodchem.2024.140886] [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: 02/27/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Fortification of human milk (HM) is often necessary to meet the nutritional requirements of preterm infants. The present experiment aimed to establish whether the supplementation of HM with either an experimental donkey milk-derived fortifier containing whole donkey milk proteins, or with a commercial bovine milk-derived fortifier containing hydrolyzed bovine whey proteins, affects peptide release differently during digestion. The experiment was conducted using an in vitro dynamic system designed to simulate the preterm infant's digestion followed by digesta analysis by means of LC-MS-MS. The different fortifiers did not appear to influence the cumulative intensity of HM peptides. Fortification had a differential impact on the release of either donkey or bovine bioactive peptides. Donkey milk peptides showed antioxidant/ACE inhibitory activities, while bovine peptides showed opioid, dipeptil- and propyl endo- peptidase inhibitory and antimicrobial activity. A slight delay in peptide release from human lactoferrin and α-lactalbumin was observed when HM was supplemented with donkey milk-derived fortifier.
Collapse
Affiliation(s)
- Marzia Giribaldi
- CNR Institute of Sciences of Food Production, Largo Braccini 2, 10095 Grugliasco, TO, Italy.
| | - Stefano Nebbia
- CNR Institute of Sciences of Food Production, Largo Braccini 2, 10095 Grugliasco, TO, Italy; INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France
| | - Valerie Briard-Bion
- INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France.
| | - Julien Jardin
- INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France.
| | - Olivia Ménard
- INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France.
| | - Didier Dupont
- INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France.
| | - Alessandra Coscia
- Neonatal Unit, University of Turin, City of Health and Science of Turin, via Ventimiglia 3, 10126 Turin, Italy.
| | - Francesco Cresi
- Neonatal Unit, University of Turin, City of Health and Science of Turin, via Ventimiglia 3, 10126 Turin, Italy.
| | - Cristina Lamberti
- CNR Institute of Sciences of Food Production, Largo Braccini 2, 10095 Grugliasco, TO, Italy.
| | - Laura Cavallarin
- CNR Institute of Sciences of Food Production, Largo Braccini 2, 10095 Grugliasco, TO, Italy.
| | - Amélie Deglaire
- INRAE, L'Institut Agro, Science et Technologie du Lait et de l'Oeuf, 65 Rue de St Brieuc, 35042 Rennes, France.
| |
Collapse
|
8
|
Jarmakiewicz-Czaja S, Sokal-Dembowska A, Ferenc K, Filip R. Mechanisms of Insulin Signaling as a Potential Therapeutic Method in Intestinal Diseases. Cells 2024; 13:1879. [PMID: 39594627 PMCID: PMC11593555 DOI: 10.3390/cells13221879] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Gastrointestinal diseases are becoming a growing public health problem. One of them is inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD). The incidence of IBD is increasing in developing countries and declining in developed countries, affecting people of all ages. Researchers have been exploring new treatment options including insulin signaling pathways in the inflammation of the gastrointestinal tract. It seems that a better understanding of the mechanism of IGF-1, GLP-1 and TL1A on the gut microbiota and inflammation may provide new advances in future therapeutic strategies for patients with IBD, but also other intestinal diseases. This review aims to synthesize insights into the effects of GLP, IGF and anti-TL1A on inflammation and the gut microbiota, which may enable their future use in therapy for people with intestinal diseases.
Collapse
Affiliation(s)
- Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
9
|
Manzoni P, Messina A, Germano C, Picone S, Masturzo B, Sainaghi PP, Sola D, Rizzi M. Lactoferrin Supplementation in Preventing and Protecting from SARS-CoV-2 Infection: Is There Any Role in General and Special Populations? An Updated Review of Literature. Int J Mol Sci 2024; 25:10248. [PMID: 39408576 PMCID: PMC11476995 DOI: 10.3390/ijms251910248] [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: 07/18/2024] [Revised: 09/07/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
At the beginning of the pandemic, SARS-CoV-2 infection represented a great medical burden worldwide, as targeted and effective therapeutic options were lacking. This resulted in the revival of existing molecules and the increasing popularity of over-the-counter nutritional supplements. Among the latter, lactoferrin has been investigated as an adjuvant in COVID-19 therapy with conflicting results, mainly depending on different study designs. Considering that lactoferrin is one of the main components of human breast milk with anti-microbial and anti-inflammatory activity, it is conceivable that such bioactive molecule could be effective in supporting anti-SARS-CoV-2 infection therapy, especially in infants and pregnant women, two subpopulations that have been poorly evaluated in the existing clinical trials. This narrative review is intended to offer insight into the existing literature on lactoferrin's biological functions and protective effects against COVID-19, with a special focus on pregnant women and their infants.
Collapse
Affiliation(s)
- Paolo Manzoni
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Alessandro Messina
- School of Medicine, University of Turin, 10124 Turin, Italy;
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Chiara Germano
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Simonetta Picone
- Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino, 00169 Rome, Italy
| | - Bianca Masturzo
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Pier Paolo Sainaghi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Daniele Sola
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Laboratory of Metabolic Research, IRCCS Istituto Auxologico Italiano, S. Giuseppe Hospital, 28824 Piancavallo, Italy
| | - Manuela Rizzi
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Department of Health Sciences (DiSS), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
10
|
Kempinski R, Arabasz D, Neubauer K. Effects of Milk and Dairy on the Risk and Course of Inflammatory Bowel Disease versus Patients' Dietary Beliefs and Practices: A Systematic Review. Nutrients 2024; 16:2555. [PMID: 39125433 PMCID: PMC11313810 DOI: 10.3390/nu16152555] [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: 07/01/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The role of the environment in the pathogenesis of inflammatory bowel disease (IBD) is undisputed, especially in light of numerous epidemiological data showing the increasing prevalence of IBD worldwide. Although no specific environmental factors have been identified, the diet has received the most attention as a potential modifier of the onset and course of IBD and as a therapeutic intervention. The Westernization of the diet is repeatedly cited as a crucial aspect of the change in IBD prevalence, but data on the impact of diet on the course of IBD are still limited and the effectiveness of dietary interventions remains uncertain. Milk remains one of the most discussed dietary agents in IBD. MATERIALS AND METHODS We performed a systematic review of the literature published between January 2010 and March 2024 on three databases, Pubmed, Web of Knowledge, and Embase, to assess the impact of milk and dairy products on the risk and course of IBD, as well as patients' dietary beliefs and practices. RESULTS We included 37 original studies in our review. CONCLUSIONS There is no clear evidence that milk and dairy products influence the incidence and course of IBD. The studies that assess this issue are characterized by great heterogeneity. Milk and dairy are among the most commonly excluded foods by patients with IBD, which may have clinical implications.
Collapse
Affiliation(s)
| | | | - Katarzyna Neubauer
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (R.K.); (D.A.)
| |
Collapse
|
11
|
Ye R, Zheng H, Yang D, Lin J, Li L, Li Y, Pan H, Dai H, Zhao L, Zhou Y, Han S, Lu Y. irAE-colitis induced by CTLA-4 and PD-1 blocking were ameliorated by TNF blocking and modulation of gut microbial. Biomed Pharmacother 2024; 177:116999. [PMID: 38925021 DOI: 10.1016/j.biopha.2024.116999] [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: 05/11/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Immune-related adverse events, particularly colitis (irAE-colitis), are significant impediments to the advancement of immune checkpoint therapy. To address this, blocking TNF-α and modulating gut microbiota are effective strategies. However, their precise roles in irAE-colitis pathogenesis and potential reciprocal relationship remain unclear. An irAE-colitis model was established to evaluate the toxicity of DICB and the efficacy of Infliximab, validated through a tumor irAE-colitis mice model. Co-administration of Infliximab with DICB mitigates colitis and enhances efficacy. Analysis of fecal samples from mice reveals altered gut microbiota composition and function induced by irAE-colitis, restored by Infliximab. Notably, Bacteriodes abundance is significantly higher in irAE-colitis. Disruption of arachidonic acid and tyrosine metabolism, and steroid hormone biosynthesis is evident. Mechanistically, a regenerative feedback loop involving DICB, TNF-α and gut microbiota underlies irAE-colitis pathogenesis. In conclusion, Infliximab shows therapeutic effects against DICB toxicity, highlighting the unforeseen roles of gut microbiota and TNF-α in irAE-colitis.
Collapse
Affiliation(s)
- Ruiwei Ye
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China; Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zheng
- Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China
| | - Dandan Yang
- Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China
| | - Jiayi Lin
- Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China
| | - Linxue Li
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China
| | - Yingying Li
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China
| | - Hanyu Pan
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China
| | - Haorui Dai
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China
| | - Liang Zhao
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China.
| | - Yonghong Zhou
- Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Sheng Han
- Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Yiming Lu
- Shanghai Baoshan Luodian Hospital, School of Medicine, Shanghai University, Shanghai 201908, China; Shanghai Marine Medical Engineering Integration Innovation Center,School of Medicine, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
12
|
Gori A, Brindisi G, Daglia M, del Giudice MM, Dinardo G, Di Minno A, Drago L, Indolfi C, Naso M, Trincianti C, Tondina E, Brunese FP, Ullah H, Varricchio A, Ciprandi G, Zicari AM. Exploring the Role of Lactoferrin in Managing Allergic Airway Diseases among Children: Unrevealing a Potential Breakthrough. Nutrients 2024; 16:1906. [PMID: 38931261 PMCID: PMC11206375 DOI: 10.3390/nu16121906] [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: 05/01/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of allergic diseases has dramatically increased among children in recent decades. These conditions significantly impact the quality of life of allergic children and their families. Lactoferrin, a multifunctional glycoprotein found in various biological fluids, is emerging as a promising immunomodulatory agent that can potentially alleviate allergic diseases in children. Lactoferrin's multifaceted properties make it a compelling candidate for managing these conditions. Firstly, lactoferrin exhibits potent anti-inflammatory and antioxidant activities, which can mitigate the chronic inflammation characteristic of allergic diseases. Secondly, its iron-binding capabilities may help regulate the iron balance in allergic children, potentially influencing the severity of their symptoms. Lactoferrin also demonstrates antimicrobial properties, making it beneficial in preventing secondary infections often associated with respiratory allergies. Furthermore, its ability to modulate the immune response and regulate inflammatory pathways suggests its potential as an immune-balancing agent. This review of the current literature emphasises the need for further research to elucidate the precise roles of lactoferrin in allergic diseases. Harnessing the immunomodulatory potential of lactoferrin could provide a novel add-on approach to managing allergic diseases in children, offering hope for improved outcomes and an enhanced quality of life for paediatric patients and their families. As lactoferrin continues to capture the attention of researchers, its properties and diverse applications make it an intriguing subject of study with a rich history and a promising future.
Collapse
Affiliation(s)
- Alessandra Gori
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| | - Giulia Brindisi
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Michele Miraglia del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Giulio Dinardo
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Alessandro Di Minno
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Lorenzo Drago
- Laboratory of Clinical Microbiology & Microbiome, Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy;
- UOC Laboratory of Clinical Medicine, MultiLab Department, IRCCS Multimedica, 20138 Milan, Italy
| | - Cristiana Indolfi
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Matteo Naso
- Allergy Center, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.N.); (C.T.)
| | - Chiara Trincianti
- Allergy Center, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.N.); (C.T.)
| | - Enrico Tondina
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | | | - Hammad Ullah
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
| | - Attilio Varricchio
- Department of Otolaryngology, University of Molise, 86100 Campobasso, Italy;
| | - Giorgio Ciprandi
- Allergy Clinic, Casa di Cura Villa Montallegro, 16145 Genoa, Italy;
| | - Anna Maria Zicari
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| |
Collapse
|
13
|
Chu X, Chen X, Zhang H, Wang Y, Guo H, Chen Y, Liu X, Zhu Z, He Y, Ding X, Wang Q, Zheng C, Cao X, Yang H, Qian J. Association of diet and outdoor time with inflammatory bowel disease: a multicenter case-control study using propensity matching analysis in China. Front Public Health 2024; 12:1368401. [PMID: 38952728 PMCID: PMC11215971 DOI: 10.3389/fpubh.2024.1368401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/09/2024] [Indexed: 07/03/2024] Open
Abstract
Objective To investigate the association between dietary and some other environmental factors and the risk of inflammatory bowel diseases (IBD) in Chinese population. Materials and methods A multicenter case-control study was conducted involving 11 hospitals across China. A total of 1,230 subjects were enrolled consecutively, and diet and environmental factor questionnaires were collected. IBD patients were matched with healthy controls (HC) using propensity-score matching (PSM) at a 1:1 ratio with a caliper value of 0.02. Multivariate conditional logistic regression analyses were performed to evaluate the associations between diet, environmental factors, and IBD. Results Moderate alcohol and milk consumption, as well as daily intake of fresh fruit, were protective factors for both Crohn's disease (CD) and ulcerative colitis (UC). Conversely, the consumption of eggs and chocolate increased the risk of IBD. Outdoor time for more than 25% of the day was a protective factor only for CD. In eastern regions of China, CD patients had higher egg consumption and less outdoor time, while UC patients consumed more chocolate. IBD patients from urban areas or with higher per capita monthly income consumed more fruit, eggs, and chocolate. Conclusions This study reveals an association between specific foods, outdoor time, and the emergence of IBD in the Chinese population. The findings emphasize the importance of a balanced diet, sufficient outdoor time and activities, and tailored prevention strategies considering regional variations.
Collapse
Affiliation(s)
- Xiaotian Chu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuanfu Chen
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huimin Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yufang Wang
- Department of Gastroenterology, Sichuan University West China Hospital, Chengdu, China
| | - Hong Guo
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yan Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yao He
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xueli Ding
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qunying Wang
- Gastroenterology and Hepatology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Changqing Zheng
- Department of Gastroenterology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaming Qian
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Sun Y, Ding Y, Liu B, Guo J, Su Y, Yang X, Man C, Zhang Y, Jiang Y. Recent advances in the bovine β-casein gene mutants on functional characteristics and nutritional health of dairy products: Status, challenges, and prospects. Food Chem 2024; 443:138510. [PMID: 38281416 DOI: 10.1016/j.foodchem.2024.138510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/04/2024] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
β-casein is the second most abundant form of casein in milk. Changes in amino acid sequence at specific positions in the primary structure of β-casein in milk will produce gene mutations that affect the physicochemical properties of dairy products and the hydrolysis site of digestive enzymes. The screening method of β-casein allele frequency detection in dairy products also has attracted the extensive attention of scientists and farmers. The A1 and A2 β-casein is the two usual mutation types, distinguished by histidine and proline at position 67 in the peptide chain. This paper summarizes the effects of A1 and A2 β-casein on the physicochemical properties of dairy products and evaluates the effects on human health, and the genotyping methods were also concluded. Impressively, this review presents possible future opportunities and challenges for the promising field of A2 β-casein, providing a valuable reference for the development of the functional dairy market.
Collapse
Affiliation(s)
- Yilin Sun
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yixin Ding
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Biqi Liu
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jinfeng Guo
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yue Su
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yu Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin 150030, China; Food Laboratory of Zhongyuan, Luohe, Henan 462300, China.
| |
Collapse
|
15
|
Liang H, Tao S, Wang Y, Zhao J, Yan C, Wu Y, Liu N, Qin Y. Astragalus polysaccharide: implication for intestinal barrier, anti-inflammation, and animal production. Front Nutr 2024; 11:1364739. [PMID: 38757131 PMCID: PMC11096541 DOI: 10.3389/fnut.2024.1364739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Intestine is responsible for nutrients absorption and plays a key role in defending against various dietary allergens, antigens, toxins, and pathogens. Accumulating evidence reported a critical role of intestine in maintaining animal and human health. Since the use of antibiotics as growth promoters in animal feed has been restricted in many countries, alternatives to antibiotics have been globally investigated, and polysaccharides are considered as environmentally friendly and promising alternatives to improve intestinal health, which has become a research hotspot due to its antibiotic substitution effect. Astragalus polysaccharide (APS), a biological macromolecule, is extracted from astragalus and has been reported to exhibit complex biological activities involved in intestinal barrier integrity maintenance, intestinal microbiota regulation, short-chain fatty acids (SCFAs) production, and immune response regulation, which are critical for intestine health. The biological activity of APS is related to its chemical structure. In this review, we outlined the source and structure of APS, highlighted recent findings on the regulation of APS on physical barrier, biochemical barrier, immunological barrier, and immune response as well as the latest progress of APS as an antibiotic substitute in animal production. We hope this review could provide scientific basis and new insights for the application of APS in nutrition, clinical medicine and health by understanding particular effects of APS on intestine health, anti-inflammation, and animal production.
Collapse
Affiliation(s)
- Hui Liang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Siming Tao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yanya Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Jing Zhao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Chang Yan
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| |
Collapse
|
16
|
Wang Y, Gong Y, Farid MS, Zhao C. Milk: A Natural Guardian for the Gut Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8285-8303. [PMID: 38588092 DOI: 10.1021/acs.jafc.3c06861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
The gut barrier plays an important role in health maintenance by preventing the invasion of dietary pathogens and toxins. Disruption of the gut barrier can cause severe intestinal inflammation. As a natural source, milk is enriched with many active constituents that contribute to numerous beneficial functions, including immune regulation. These components collectively serve as a shield for the gut barrier, protecting against various threats such as biological, chemical, mechanical, and immunological threats. This comprehensive review delves into the active ingredients in milk, encompassing casein, α-lactalbumin, β-lactoglobulin, lactoferrin, the milk fat globular membrane, lactose, transforming growth factor, and glycopeptides. The primary focus is to elucidate their impact on the integrity and function of the gut barrier. Furthermore, the implications of different processing methods of dairy products on the gut barrier protection are discussed. In conclusion, this study aimed to underscore the vital role of milk and dairy products in sustaining gut barrier health, potentially contributing to broader perspectives in nutritional sciences and public health.
Collapse
Affiliation(s)
- Yanli Wang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yiyao Gong
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | | | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| |
Collapse
|
17
|
Li B, Zhang B, Zhang F, Liu X, Zhang Y, Peng W, Teng D, Mao R, Yang N, Hao Y, Wang J. Interaction between Dietary Lactoferrin and Gut Microbiota in Host Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7596-7606. [PMID: 38557058 DOI: 10.1021/acs.jafc.3c09050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The gut microbiota are known to play an important role in host health and disease. Alterations in the gut microbiota composition can disrupt the stability of the gut ecosystem, which may result in noncommunicable chronic diseases (NCCDs). Remodeling the gut microbiota through personalized nutrition is a novel therapeutic avenue for both disease control and prevention. However, whether there are commonly used gut microbiota-targeted diets and how gut microbiota-diet interactions combat NCCDs and improve health remain questions to be addressed. Lactoferrin (LF), which is broadly used in dietary supplements, acts not only as an antimicrobial in the defense against enteropathogenic bacteria but also as a prebiotic to propagate certain probiotics. Thus, LF-induced gut microbiota alterations can be harnessed to induce changes in host physiology, and the underpinnings of their relationships and mechanisms are beginning to unravel in studies involving humans and animal models.
Collapse
Affiliation(s)
- Bing Li
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Bo Zhang
- International Joint Research Laboratory for Biomedical Nanomaterials of Henan, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Fuli Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Xiaomeng Liu
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Yunxia Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Weifeng Peng
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Da Teng
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Ruoyu Mao
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Na Yang
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Ya Hao
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| | - Jianhua Wang
- Gene Engineering Lab, Feed Research Institute, Chinese Academy of Agricultural Science, Beijing 100081, P. R. China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, P. R. China
| |
Collapse
|
18
|
Paziewska-Nowak A, Urbanowicz M, Sadowska K, Pijanowska DG. DNA-based molecular recognition system for lactoferrin biosensing. Int J Biol Macromol 2023; 253:126747. [PMID: 37699464 DOI: 10.1016/j.ijbiomac.2023.126747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
The paper describes the development of a novel DNA oligonucleotide-based affinity bioreceptor that binds to lactoferrin, a glycoprotein-type immunomodulator. The research was performed using surface plasmon resonance method to investigate affinity of various types of oligonucleotides to the target protein. The 72 base pair-long 5'[(TAGAGGATCAAA)AAA]4TAGAGGATCAAA3' sequence with the highest affinity to lactoferrin was selected for further investigations. Kinetic analysis of the interaction between selected DNA and lactoferrin provided rate and equilibrium constants: ka = (2.49 ± 0.03)∙104 M-1∙s-1, kd = (1.89 ± 0.02)∙10-3 s-1, KA = (0.13 ± 0.05)∙108 M-1, and KD = (7.61 ± 0.18)∙10-8 M. Thermodynamic study conducted to determine the ΔH0, ΔS0, and ΔG0 for van't Hoff characteristic in the temperature range of 291.15-305.15 K, revealed the complex formation as endothermic and entropically driven. The chosen DNA sequence's selectivity towards lactoferrin was confirmed with interferents' response constituting <3 % of the response to lactoferrin. SPR analysis justified utility of the designed DNA oligonucleotide for Lf determination, with LOD of 4.42∙10-9 M. Finally, the interaction between lactoferrin and DNA was confirmed by electrochemical impedance spectroscopy, providing the basis for further quantitative assay of lactoferrin using the developed DNA-based bioreceptor. The interactions were performed under immobilized DNA ligand conditions, thus reflecting the sensor's surface, which facilitates their transfer to other label-free biosensor technologies.
Collapse
Affiliation(s)
- Agnieszka Paziewska-Nowak
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 St., 02-109 Warsaw, Poland.
| | - Marcin Urbanowicz
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 St., 02-109 Warsaw, Poland
| | - Kamila Sadowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 St., 02-109 Warsaw, Poland
| | - Dorota Genowefa Pijanowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 St., 02-109 Warsaw, Poland
| |
Collapse
|
19
|
Gad El-Hak HN, Mohamed FH. Effect of lactoferrin supplement on cadmium chloride induced toxicity to male rats: Toxicopathological, ultrastructural and immunological studies. Int Immunopharmacol 2023; 125:111182. [PMID: 37944217 DOI: 10.1016/j.intimp.2023.111182] [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: 07/22/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
This study sought to determine whether lactoferrin supplementation could counteract the harm that cadmium (Cd) induced to the rats. The effect of Cd and lactoferrin were investigated in hematological, biochemical, histological, immunohistochemical expression and ultrastructural studies. After 30 days of treatment, rats exposed to Cd had significantly higher levels of Cd in their blood, more oxidized lipids, and less antioxidant capacity overall. Supplemental lactoferrin also significantly undoes that effect. Hematological and biochemical parameters changed along with the increase in blood Cd levels. The histological integrity of the liver, kidney, spleen, and (axillary, cervical, mesenteric and popliteal) lymph nodes that had been damaged by Cd exposure was also restored by lactoferrin supplementation. Moreover, the liver and spleen ultrastructure showed the same improvement. In addition, the spleen of Lf/Cd group showed less immunohistochemical expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in comparison to the Cd group. In conclusion, the current study showed that supplementing with lactoferrin improved immune response and restored biochemical and oxidative stability induced by Cd.
Collapse
|
20
|
Sienkiewicz M, Zielińska M, Jacenik D, Machelak W, Owczarek K, Fichna J. Lactoferrin improves symptoms of dextran sulfate sodium-induced colitis in mice through modulation of cellular senescence. Nutr Res 2023; 120:58-71. [PMID: 37931351 DOI: 10.1016/j.nutres.2023.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023]
Abstract
The multifaceted effects of lactoferrin (LF) on the digestive and immune systems make it an attractive therapeutic option in inflammatory bowel diseases. In this study, we aimed to explore the anti-inflammatory effects of LF in colitis, particularly in relation to cellular senescence. We hypothesize that LF has the potential to modulate the senescence process. The effects of LF on senescence were tested in vitro using HCT116 and SW480 cell lines, and in vivo, the dextran sulfate sodium-induced mouse model of colitis. LF (500 mg/kg) alleviated symptoms of colitis in mice with a significant decrease in colon damage (P < .0001 vs. control) and microscopic (P < .05 vs. control) scores. Cellular senescence markers p16 and p21 were significantly upregulated in the mouse colon during inflammation (both P < .01 vs. control), and LF at 500 mg/kg decreased these markers (both P < .05 vs. dextran sulfate sodium-treated mice). In vitro, LF significantly affected the expression of p16 and p21 (P < .05-P < .0001 vs. control), senescence associated secretory phenotype (P < .01-P < .0001 vs. control), and telomere-specific proteins: telomeric repeat binding factor 1 and 2 (P < .05-P < .0001 vs. control) in a concentration-dependent manner. LF modulates the expression of cellular senescence markers and shows hallmarks of senolytic and pro-senescent activity, depending on dose. Further studies are needed to fully understand the anti-inflammatory effect of LF in the context of senescence and safe utilization in patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Michał Sienkiewicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Marta Zielińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| | - Weronika Machelak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Katarzyna Owczarek
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland.
| |
Collapse
|
21
|
Guiducci L, Cabiati M, Santocchi E, Prosperi M, Morales MA, Muratori F, Randazzo E, Federico G, Calderoni S, Del Ry S. Expression of miRNAs in Pre-Schoolers with Autism Spectrum Disorders Compared with Typically Developing Peers and Its Effects after Probiotic Supplementation. J Clin Med 2023; 12:7162. [PMID: 38002774 PMCID: PMC10672692 DOI: 10.3390/jcm12227162] [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: 10/25/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Alteration of the microbiota-gut-brain axis has been recently recognized as a possible contributor to the physiopathology of autism spectrum disorder (ASD). In this context, microRNA (miRNAs) dysfunction, implicated both in several neuropathological conditions including ASD and in different gastrointestinal disorders (GIDs), could represent an important modulating factor. In this contextual framework, we studied the transcriptional profile of specific circulating miRNAs associated with both ASD (miR-197-5p, miR-424-5p, miR-500a-5p, miR-664a-5p) and GID (miR-21-5p, miR-320a-5p, miR-31-5p, miR-223-5p) in a group of pre-schoolers with ASD and in typically developing (TD) peers. In the ASD group, we also assessed the same miRNAs after a 6-month supplementation with probiotics and their correlation with plasma levels of zonulin and lactoferrin. At baseline, the expression of miRNAs involved in ASD were significantly reduced in ASD pre-schoolers vs. TD controls. Regarding the miRNAs involved in GID, the expression levels of miR-320-5p, miR-31-5p, and miR-223-5p were significantly higher in ASD than in TD subjects, whereas miR-21-5p showed significantly reduced expression in the ASD group vs. TD group. Supplementation with probiotics did not significantly change the expression of miRNAs in the ASD population. We found a significative negative correlation between zonulin and miR-197-5p and miR-21-5p at baseline, as well as between lactoferrin and miR-223-5p after 6 months of probiotic supplementation. Our study confirms the presence of an altered profile of the miRNAs investigated in ASD versus TD peers that was not modified by supplementation with probiotics.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Manuela Cabiati
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Elisa Santocchi
- UFSMIA Zona Valle del Serchio, Azienda USL Toscana Nord Ovest, 55032 Castelnuovo di Garfagnana, Italy;
| | - Margherita Prosperi
- UFSMIA Valdera-Alta Val di Cecina, Azienda USL Toscana Nord Ovest, 56128 Pisa, Italy;
| | - Maria Aurora Morales
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| | - Filippo Muratori
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Emioli Randazzo
- Unit of Pediatric Endocrinology and Diabetes, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (E.R.); (G.F.)
| | - Giovanni Federico
- Unit of Pediatric Endocrinology and Diabetes, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (E.R.); (G.F.)
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Ry
- CNR, Institute of Clinical Physiology, 56124 Pisa, Italy; (L.G.); (M.C.); (M.A.M.); (S.D.R.)
| |
Collapse
|
22
|
Liu X, Xi C, Li W, Su H, Yang H, Bai Z, Tian Y, Song S. Moringa oleifera Leaves Protein Enhances Intestinal Permeability by Activating TLR4 Upstream Signaling and Disrupting Tight Junctions. Int J Mol Sci 2023; 24:16425. [PMID: 38003615 PMCID: PMC10671199 DOI: 10.3390/ijms242216425] [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: 09/12/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Changes in intestinal mucosal barrier permeability lead to antigen sensitization and mast cell-mediated allergic reactions, which are considered to play important roles in the occurrence and development of food allergies. It has been suggested that protein causes increased intestinal permeability via mast cell degranulation, and we investigated the effect of camellia Moringa oleifera leaves protein on intestinal permeability and explored its role in the development of food allergies. The current study investigated the effect of M. oleifera leaves protein on intestinal permeability through assessments of transepithelial electrical resistance (TEER) and transmembrane transport of FITC-dextran by Caco-2 cells. The expression levels of Toll-like receptor 4 (TLR4), IL-8, Occludin, Claudin-1, and perimembrane protein family (ZO-1) were detected by real-time PCR and Western blotting. The effect of M. oleifera leaves protein on intestinal permeability was verified in mice in vivo. The serum fluorescence intensity was measured using the FITC-dextran tracer method, and the expression of tight junction proteins was detected using Western blotting. The results showed that M. oleifera leaves protein widened the gaps between Caco-2 cells, reduced transmembrane resistance, and increased permeability. This protein also reduced the mRNA and protein levels of Occludin, Claudin-1, and ZO-1. Animal experiments showed that intestinal permeability was increased, and that the expression of the tight junction proteins Occludin and Claudin-1 were downregulated in mice. This study shows that M. oleifera leaves protein has components that increase intestinal permeability, decrease tight junction protein expression, promote transmembrane transport in Caco-2 cells, and increase intestinal permeability in experimental animals. The finding that M. oleifera leaves active protein increases intestinal permeability suggests that this protein may be valuable for the prevention, diagnosis, and treatment of M. oleifera leaves allergy.
Collapse
Affiliation(s)
- Xiaoxue Liu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
| | - Chuyu Xi
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
| | - Wenjie Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
| | - Hairan Su
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
| | - Hao Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
| | - Zhongbin Bai
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China;
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yang Tian
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China;
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
| | - Shuang Song
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (C.X.); (W.L.); (H.S.); (H.Y.)
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China;
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
23
|
Zhao C, Chen N, Ashaolu TJ. Prebiotic and modulatory evidence of lactoferrin on gut health and function. J Funct Foods 2023; 108:105741. [DOI: 10.1016/j.jff.2023.105741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
24
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
25
|
Artym J, Zimecki M. Colostrum and Lactoferrin Protect against Side Effects of Therapy with Antibiotics, Anti-inflammatory Drugs and Steroids, and Psychophysical Stress: A Comprehensive Review. Biomedicines 2023; 11:1015. [PMID: 37189633 PMCID: PMC10136316 DOI: 10.3390/biomedicines11041015] [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: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
In this article, we review the benefits of applying bovine colostrum (BC) and lactoferrin (LF) in animal models and clinical trials that include corticosteroid application and psychic stress, treatment with non-steroid anti-inflammatory drugs (NSAIDs) and antibiotics. A majority of the reported investigations were performed with native bovine or recombinant human LF, applied alone or in combination with probiotics, as nutraceutics and diet supplements. Apart from reducing adverse side effects of the applied therapeutics, BC and LF augmented their efficacy and improved the wellness of patients. In conclusion, LF and complete native colostrum, preferably administered with probiotic bacteria, are highly recommended for inclusion in therapeutic protocols in NSAIDs and corticosteroid anti-inflammatory, as well as antibiotic, therapies. These colostrum-based products can also be of value for individuals subjected to prolonged psychophysical stress (mediated by endogenous corticosteroids), especially at high ambient temperatures (soldiers and emergency services), as well as physically active people and training athletes. They are also recommended for patients during recovery from trauma and surgery, which are always associated with severe psychophysical stress.
Collapse
Affiliation(s)
| | - Michał Zimecki
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12 Str., 53-114 Wroclaw, Poland
| |
Collapse
|
26
|
Ohradanova-Repic A, Praženicová R, Gebetsberger L, Moskalets T, Skrabana R, Cehlar O, Tajti G, Stockinger H, Leksa V. Time to Kill and Time to Heal: The Multifaceted Role of Lactoferrin and Lactoferricin in Host Defense. Pharmaceutics 2023; 15:1056. [PMID: 37111542 PMCID: PMC10146187 DOI: 10.3390/pharmaceutics15041056] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lactoferrin is an iron-binding glycoprotein present in most human exocrine fluids, particularly breast milk. Lactoferrin is also released from neutrophil granules, and its concentration increases rapidly at the site of inflammation. Immune cells of both the innate and the adaptive immune system express receptors for lactoferrin to modulate their functions in response to it. On the basis of these interactions, lactoferrin plays many roles in host defense, ranging from augmenting or calming inflammatory pathways to direct killing of pathogens. Complex biological activities of lactoferrin are determined by its ability to sequester iron and by its highly basic N-terminus, via which lactoferrin binds to a plethora of negatively charged surfaces of microorganisms and viruses, as well as to mammalian cells, both normal and cancerous. Proteolytic cleavage of lactoferrin in the digestive tract generates smaller peptides, such as N-terminally derived lactoferricin. Lactoferricin shares some of the properties of lactoferrin, but also exhibits unique characteristics and functions. In this review, we discuss the structure, functions, and potential therapeutic uses of lactoferrin, lactoferricin, and other lactoferrin-derived bioactive peptides in treating various infections and inflammatory conditions. Furthermore, we summarize clinical trials examining the effect of lactoferrin supplementation in disease treatment, with a special focus on its potential use in treating COVID-19.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Praženicová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Laura Gebetsberger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tetiana Moskalets
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Ondrej Cehlar
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Gabor Tajti
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vladimir Leksa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| |
Collapse
|
27
|
Levy E, Marcil V, Tagharist Ép Baumel S, Dahan N, Delvin E, Spahis S. Lactoferrin, Osteopontin and Lactoferrin–Osteopontin Complex: A Critical Look on Their Role in Perinatal Period and Cardiometabolic Disorders. Nutrients 2023; 15:nu15061394. [PMID: 36986124 PMCID: PMC10052990 DOI: 10.3390/nu15061394] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Milk-derived bioactive proteins have increasingly gained attention and consideration throughout the world due to their high-quality amino acids and multiple health-promoting attributes. Apparently, being at the forefront of functional foods, these bioactive proteins are also suggested as potential alternatives for the management of various complex diseases. In this review, we will focus on lactoferrin (LF) and osteopontin (OPN), two multifunctional dairy proteins, as well as to their naturally occurring bioactive LF–OPN complex. While describing their wide variety of physiological, biochemical, and nutritional functionalities, we will emphasize their specific roles in the perinatal period. Afterwards, we will evaluate their ability to control oxidative stress, inflammation, gut mucosal barrier, and intestinal microbiota in link with cardiometabolic disorders (CMD) (obesity, insulin resistance, dyslipidemia, and hypertension) and associated complications (diabetes and atherosclerosis). This review will not only attempt to highlight the mechanisms of action, but it will critically discuss the potential therapeutic applications of the underlined bioactive proteins in CMD.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Valérie Marcil
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Sarah Tagharist Ép Baumel
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Noam Dahan
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
| | - Edgard Delvin
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Schohraya Spahis
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
- Correspondence: ; Tel.: +1-(514)-345-4832
| |
Collapse
|
28
|
Einerhand AWC, van Loo-Bouwman CA, Weiss GA, Wang C, Ba G, Fan Q, He B, Smit G. Can Lactoferrin, a Natural Mammalian Milk Protein, Assist in the Battle against COVID-19? Nutrients 2022; 14:nu14245274. [PMID: 36558432 PMCID: PMC9782828 DOI: 10.3390/nu14245274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Notwithstanding mass vaccination against specific SARS-CoV-2 variants, there is still a demand for complementary nutritional intervention strategies to fight COVID-19. The bovine milk protein lactoferrin (LF) has attracted interest of nutraceutical, food and dairy industries for its numerous properties-ranging from anti-viral and anti-microbial to immunological-making it a potential functional ingredient in a wide variety of food applications to maintain health. Importantly, bovine LF was found to exert anti-viral activities against several types of viruses, including certain SARS-CoV-2 variants. LF's potential effect on COVID-19 patients has seen a rapid increase of in vitro and in vivo studies published, resulting in a model on how LF might play a role during different phases of SARS-CoV-2 infection. Aim of this narrative review is two-fold: (1) to highlight the most relevant findings concerning LF's anti-viral, anti-microbial, iron-binding, immunomodulatory, microbiota-modulatory and intestinal barrier properties that support health of the two most affected organs in COVID-19 patients (lungs and gut), and (2) to explore the possible underlying mechanisms governing its mode of action. Thanks to its potential effects on health, bovine LF can be considered a good candidate for nutritional interventions counteracting SARS-CoV-2 infection and related COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Caiyun Wang
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
| | - Genna Ba
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Qicheng Fan
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Baoping He
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Gerrit Smit
- Yili Innovation Center Europe, 6708 WH Wageningen, The Netherlands
| |
Collapse
|
29
|
Li C, Liu X, Huang Z, Zhai Y, Li H, Wu J. Lactoferrin Alleviates Lipopolysaccharide-Induced Infantile Intestinal Immune Barrier Damage by Regulating an ELAVL1-Related Signaling Pathway. Int J Mol Sci 2022; 23:13719. [PMID: 36430202 PMCID: PMC9696789 DOI: 10.3390/ijms232213719] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
As the most important intestinal mucosal barrier of the main body, the innate immune barrier in intestinal tract plays especially pivotal roles in the overall health conditions of infants and young children; therefore, how to strengthen the innate immune barrier is pivotal. A variety of bioactivities of lactoferrin (LF) has been widely proved, including alleviating enteritis and inhibiting colon cancer; however, the effects of LF on intestinal immune barrier in infants and young children are still unclear, and the specific mechanism on how LF inhibits infantile enteritis by regulating immune signaling pathways is unrevealed. In the present study, we firstly performed pharmacokinetic analyses of LF in mice intestinal tissues, stomach tissues and blood, through different administration methods, to confirm the metabolic method of LF in mammals. Then we constructed in Vitro and in Vivo infantile intestinal immune barrier damage models utilizing lipopolysaccharide (LPS), and evaluated the effects of LF in alleviating LPS-induced intestinal immune barrier damage. Next, the related immune molecular mechanism on how LF exerted protective effects was investigated, through RNA-seq analyses of the mouse primary intestinal epithelial cells, and the specific genes were analyzed and screened out. Finally, the genes and their related immune pathway were validated in mRNA and protein levels; the portions of special immune cells (CD4+ T cells and CD8+ T cells) were also detected to further support our experimental results. Pharmacokinetic analyses demonstrated that the integrity of LF could reach mice stomach and intestine after oral gavage within 12 h, and the proper administration of LF should be the oral route. LF was proven to down-regulate the expression levels of inflammatory cytokines in both the primary intestinal epithelial cells and mice blood, especially LF without iron (Apo-LF), indicating LF alleviated infantile intestinal immune barrier damage induced by LPS. And through RNA-seq analyses of the mouse primary intestinal epithelial cells treated with LPS and LF, embryonic lethal abnormal vision Drosophila 1 (ELAVL1) was selected as one of the key genes, then the ELAVL1/PI3K/NF-κB pathway regulated by LF was verified to participate in the protection of infantile intestinal immune barrier damage in our study. Additionally, the ratio of blood CD4+/CD8+ T cells was significantly higher in the LF-treated mice than in the control mice, indicating that LF distinctly reinforced the overall immunity of infantile mice, further validating the strengthening bioactivity of LF on infantile intestinal immune barrier. In summary, LF was proven to alleviate LPS-induced intestinal immune barrier damage in young mice through regulating ELAVL1-related immune signaling pathways, which would expand current knowledge of the functions of bioactive proteins in foods within different research layers, as well as benefit preclinical and clinical researches in a long run.
Collapse
Affiliation(s)
- Chaonan Li
- Beijing Key Laboratory of Food Processing and Safety in Forestry, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100085, China
| | - Xinkui Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihong Huang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yiyan Zhai
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huiying Li
- Beijing Key Laboratory of Food Processing and Safety in Forestry, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100085, China
| | - Jiarui Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
30
|
Li DM, Wu YX, Hu ZQ, Wang TC, Zhang LL, Zhou Y, Tong X, Xu JY, Qin LQ. Lactoferrin Prevents Chronic Alcoholic Injury by Regulating Redox Balance and Lipid Metabolism in Female C57BL/6J Mice. Antioxidants (Basel) 2022; 11:antiox11081508. [PMID: 36009227 PMCID: PMC9405310 DOI: 10.3390/antiox11081508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the preventive effects of lactoferrin (Lf) on chronic alcoholic liver injury (ALI) in female mice. Female C57BL/6J mice were randomly divided into four groups: control group (CON), ethanol administration group (EtOH), low-dose Lf treatment group (LLf), and high-dose Lf group (HLf). In the last three groups, chronic ALI was induced by administering 20% ethanol ad libitum for 12 weeks. Mice in the CON and EtOH groups were fed with AIN-93G diet. Meanwhile, 0.4% and 4% casein in the AIN-93G diet were replaced by Lf as the diets of LLf and HLf groups, respectively. HLf significantly reduced hepatic triglyceride content and improved pathological morphology. HLf could inhibit cytochrome P450 2E1 overexpression and promote alcohol dehydrogenase-1 expression. HLf activated protein kinase B and AMP-activated protein kinase (AMPK), as well as upregulating nuclear-factor-erythroid-2-related factor-2 expression to elevate hepatic antioxidative enzyme activities. AMPK activation also benefited hepatic lipid metabolism. Meanwhile, HLf had no obvious beneficial effects on gut microbiota. In summary, Lf could alleviate chronic ALI in female mice, which was associated with redox balance and lipid metabolism regulation.
Collapse
Affiliation(s)
- De-Ming Li
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Yun-Xuan Wu
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Zhi-Qiang Hu
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Tian-Ci Wang
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Li-Li Zhang
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Yan Zhou
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
| | - Xing Tong
- Laboratory Center, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China;
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Correspondence: (J.-Y.X.); (L.-Q.Q.)
| | - Li-Qiang Qin
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China; (D.-M.L.); (Y.-X.W.); (Z.-Q.H.); (T.-C.W.); (L.-L.Z.); (Y.Z.)
- Correspondence: (J.-Y.X.); (L.-Q.Q.)
| |
Collapse
|